US11555038B2 - Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof - Google Patents

Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof Download PDF

Info

Publication number
US11555038B2
US11555038B2 US16/479,709 US201816479709A US11555038B2 US 11555038 B2 US11555038 B2 US 11555038B2 US 201816479709 A US201816479709 A US 201816479709A US 11555038 B2 US11555038 B2 US 11555038B2
Authority
US
United States
Prior art keywords
crystalline form
compound
solvent
heptane
etoac
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US16/479,709
Other versions
US20210332049A1 (en
Inventor
Yunhang Guo
Desheng YU
Zhiwei Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beigene Ltd
Original Assignee
Beigene Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beigene Ltd filed Critical Beigene Ltd
Assigned to BEIGENE, LTD. reassignment BEIGENE, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WANG, ZHIWEI, GUO, Yunhang, YU, Desheng
Publication of US20210332049A1 publication Critical patent/US20210332049A1/en
Application granted granted Critical
Publication of US11555038B2 publication Critical patent/US11555038B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • Compound 1 Disclosed herein are crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide (hereinafter Compound 1); methods for preparing crystalline forms of Compound 1; pharmaceutical compositions comprising crystalline forms of Compound 1 and a pharmaceutically acceptable carrier; and methods of using crystalline forms of Compound 1 as a Btk inhibitor for treating or preventing diseases.
  • Compound 1 crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Btk Bruton's tyrosine kinase belongs to the Tec tyrosine kinase family (Vetrie et al., Nature 361: 226-233, 1993; Bradshaw, Cell Signal. 22: 1175-84, 2010). Btk is primarily expressed in most hematopoietic cells such as B cells, mast cells and macrophages (Smith et al., J. Immunol. 152: 557-565, 1994) and is localized in bone marrow, spleen and lymph node tissue. Btk plays important roles in B-cell receptor (BCR) and FcR signaling pathways, which involve in B-cell development, differentiation (Khan, Immunol. Res.
  • BCR B-cell receptor
  • FcR FcR signaling pathways
  • Btk is activated by upstream Src-family kinases. Once activated, Btk in turn phosphorylates PLC gamma, leading to effects on B-cell function and survival (Humphries et al., J Biol. Chem. 279: 37651, 2004).
  • Btk inhibitors can be used to treat autoimmune and/or inflammatory diseases.
  • Btk inhibitor PCI-32765 was effective in treatment of several types of B-cell lymphoma (for example, 54th American Society of Hematology (ASH) annual meeting abstract, December 2012: 686
  • Btk Bruton's Tyrosine Kinase
  • Ibrutinib PCI-32765
  • DLBCL ABC Subtype of Relapsed/Refractory De Novo Diffuse Large B-Cell Lymphoma
  • Btk plays a central role as a mediator in multiple signal transduction pathways, inhibitors of Btk are of great interest as anti-inflammatory and/or anti-cancer agents (Mohamed et al., Immunol. Rev. 228: 58-73, 2009; Pan, Drug News perspect 21: 357-362, 2008; Rokosz et al., Expert Opin. Ther. Targets 12: 883-903, 2008; Uckun et al., Anti - cancer Agents Med. Chem. 7: 624-632, 2007; Lou et al, J. Med. Chem. 55(10): 4539-4550, 2012).
  • WO2014173289A disclosed a series of fused heterocyclic compounds as Btk inhibitors.
  • WO2014173289A disclosed (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Compound 1 is a potent, specific and irreversible BTK kinase inhibitor.
  • the data generated in preclinical studies using biochemical, cell based and animal studies have suggested that Compound 1 is of significant benefit in inhibiting tumor growth in B-cell malignancies.
  • Compound 1 was shown to be more selective than ibrutinib for inhibition of BTK vs. EGFR, FGR, FRK, HER2, HER4, ITK, JAK3, LCK, and TEC, indicating that Compound 1 will have less side-effects than ibrutinib in clinic.
  • Compound 1 prepared in accordance with WO2014173289A was found to be in amorphous form, as confirmed by the X-Ray Powder Diffraction (hereinafter XRPD) pattern of FIG. 1 A (the existence of a large bump distributed in a wide range) and the TGA/mDSC curves of FIG. 1 B (no substantial glass transition signal).
  • XRPD X-Ray Powder Diffraction
  • the amorphous form of Compound 1 presents some challenges for drug formulation due to its low stability and difficulties to handle. Therefore, there is a need to develop new forms of Compound 1, especially crystalline forms, which are stable and easy to handle and to process in the manufacture and preparation of drug formulations.
  • the inventors of the present invention have found solvated or non-solvated crystalline forms of Compound 1, which are stable and easy to handle and to process in the manufacture and preparation of drug formulations compared with the amorphous form of Compound 1.
  • an anhydrous non-solvated crystalline form i.e., Form B herein
  • Form B anhydrous non-solvated crystalline form
  • the crystalline form of Compound 1 is solvated (herein referred to as “Form A” or “Form A as a solvate” or “solvated crystalline form”).
  • the crystalline form of Compound 1 is anhydrous non-solvated (herein referred to as “Form B” or “non-solvated crystalline form”).
  • a pharmaceutical composition comprising a therapeutically effective amount of Form A or B disclosed herein and a pharmaceutically acceptable carrier.
  • the disease is an allergic disease, an autoimmune disease, an inflammatory disease, or a cancer, or a combination of two or more thereof.
  • the disease is a B-cell proliferative disease, selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, or chronic lymphocytic leukemia, or a combination of two or more thereof.
  • the inflammatory disease is selected from rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis.
  • the disease is an allergic disease, an autoimmune disease, an inflammatory disease, or a cancer, or a combination of two or more thereof.
  • the disease is a B-cell proliferative disease, selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, or chronic lymphocytic leukemia, or a combination of two or more thereof.
  • the inflammatory disease is selected from rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis.
  • FIG. 1 A shows the XRPD pattern of the amorphous form of Compound 1.
  • FIG. 1 B shows the TGA/mDSC curves of amorphous form of Compound 1.
  • FIG. 2 A shows the XRPD pattern of Form A (crystallization from EtOAc/n-heptane).
  • FIG. 2 B shows the TGA/DSC curves of Form A (crystallization from EtOAc/n-heptane).
  • FIG. 2 C shows the 1 H-NMR of Form A (crystallization from EtOAc/n-heptane).
  • FIG. 2 D shows XRPD overlay of Form A (crystallization from EtOAc/n-heptane) before and after heating.
  • FIG. 3 A shows the XRPD pattern of Form A (crystallization from EtOH/n-heptane) and Form A (crystallization from acetone/n-heptane).
  • FIG. 3 B shows the XRPD pattern of Form A obtained from slurry conversion of EtOH/n-heptane.
  • FIG. 3 C shows the TGA curve of Form A obtained from slurry conversion of EtOH/n-heptane.
  • FIG. 3 D shows the 1 H-NMR of Form A obtained from slurry conversion of EtOH/n-heptane.
  • FIG. 3 E shows the XRPD overlay of Form A obtained from slurry conversion of EtOH/n-heptane before and after heating.
  • FIG. 4 A shows the XRPD pattern of Crystalline Form B.
  • FIG. 4 B shows the TGA/DSC curves of Form B.
  • FIG. 4 C shows the 1 H-NMR of Crystalline Form B.
  • FIG. 4 D shows the 13 C-NMR of Crystalline Form B.
  • FIG. 4 E shows DVS plot of Crystalline Form B.
  • FIG. 4 F shows XRPD patterns of Form B pre and post DVS.
  • FIG. 4 G shows XRPD overlay of Form B of Example 4, Example 5A and Example 5B.
  • FIG. 4 H shows XRPD overlay of Form B before and after grinding.
  • FIG. 5 A shows XRPD overlay for slurry at RT starting from amorphous Compound 1.
  • FIG. 5 B shows XRPD overlay for slurry at 50° C. starting from amorphous Compound 1.
  • FIG. 5 C shows XRPD overlay for competitive slurry of Form A and B at 10° C.
  • FIG. 5 D shows XRPD overlay for competitive slurry of Form A and B at RT.
  • FIG. 5 E shows XRPD overlay for competitive slurry of Form A and B at 50° C.
  • FIG. 6 A shows the absolute structure of single crystal of BG-13.
  • FIG. 6 B illustrates hydrogen bonds of single crystal of BG-13.
  • FIG. 6 C shows a crystal packing of single crystal of BG-13.
  • FIG. 6 D shows the XRPD pattern of single crystal of BG-13.
  • Compound 1 can exist in crystalline forms, either solvated or non-solvated.
  • Compound 1 is in solvated crystalline form (i.e., Form A).
  • Form A is solvated with EtOAc (also referred to as “Form A as an EtOAc solvate”).
  • Form A is solvated with other solvent or anti-solvent or is to form a hetero-solvate.
  • Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 5.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 18.6 ⁇ 0.2°, 19.8 ⁇ 0.2° and 20.6 ⁇ 0.2°.
  • Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 5.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 18.6 ⁇ 0.2°, 19.8 ⁇ 0.2° and 20.6 ⁇ 0.2°.
  • Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 5.3 ⁇ 0.2°, 10.9 ⁇ 0.2°, 16.9 ⁇ 0.2°, 18.6 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.1 ⁇ 0.2°, 22.5 ⁇ 0.2° and 22.8 ⁇ 0.20.
  • Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 5.3 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.2 ⁇ 0.2°, 13.4 ⁇ 0.2°, 14.4 ⁇ 0.2°, 16.9 ⁇ 0.2°, 18.6 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.1 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.5 ⁇ 0.2°, 22.8 ⁇ 0.2°, 23.6 ⁇ 0.2° and 24.3 ⁇ 0.2°.
  • Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 5.3 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.2 ⁇ 0.2°, 12.0 ⁇ 0.2°, 13.4 ⁇ 0.2°, 14.1 ⁇ 0.2°, 14.4 ⁇ 0.2°, 16.9 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.6 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.1 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.5 ⁇ 0.2°, 22.8 ⁇ 0.2°, 23.6 ⁇ 0.2°, 24.0 ⁇ 0.2° and 24.3 ⁇ 0.2°.
  • Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 5.3 ⁇ 0.2°, 9.2 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.2 ⁇ 0.2°, 12.0 ⁇ 0.2°, 13.4 ⁇ 0.2°, 14.1 ⁇ 0.2°, 14.4 ⁇ 0.2°, 15.5 ⁇ 0.2°, 16.9 ⁇ 0.2°, 17.7 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.6 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.1 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.5 ⁇ 0.2°, 22.8 ⁇ 0.2°, 23.6 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.3 ⁇ 0.2° and 26.4 ⁇ 0.2°.
  • Compound 1 is in Form A characterized by an XRPD substantially in accordance with FIG. 2 A .
  • Compound 1 is in Form A characterized by an XRPD summarized in Table 1.
  • Compound 1 is in Form A as an EtOAc solvate characterized by a DSC substantially in accordance with FIG. 2 B .
  • Compound 1 is in Form A as an EtOAc solvate characterized by a TGA substantially in accordance with FIG. 2 B .
  • Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 15.1 ⁇ 0.2°, 20.7 ⁇ 0.2° and 22.8 ⁇ 0.2°.
  • Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 15.1 ⁇ 0.2°, 15.5 ⁇ 0.2°, 19.9 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.7 ⁇ 0.2° and 22.8 ⁇ 0.2°.
  • Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 15.1 ⁇ 0.2°, 15.5 ⁇ 0.2°, 17.0 ⁇ 0.2°, 19.9 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.8 ⁇ 0.2° and 24.2 ⁇ 0.20.
  • Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 6.3 ⁇ 0 0.2°, 9.5 ⁇ 0.2°, 15.1 ⁇ 0.2°, 15.5 ⁇ 0.2°, 16.5 ⁇ 0.2°, 17.0 ⁇ 0.2°, 19.9 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.8 ⁇ 0.2°, 24.2 ⁇ 0.2° and 25.1 ⁇ 0.2°.
  • Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2 ⁇ angle values independently selected from: approximately 6.3 ⁇ 0 0.2°, 9.5 ⁇ 0.2°, 12.4 ⁇ 0.2°, 13.2 ⁇ 0.2°, 15.1 ⁇ 0.2°, 15.5 ⁇ 0.2°, 16.5 ⁇ 0.2°, 17.0 ⁇ 0.2°, 18.0 ⁇ 0.2°, 18.9 ⁇ 0.2°, 19.9 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.8 ⁇ 0.2°, 24.2 ⁇ 0.2°, 25.1 ⁇ 0.2°, 25.8 ⁇ 0.2°, 26.7 ⁇ 0.2° and 27.6 ⁇ 0.2°.
  • Compound 1 is in Form B characterized by an XRPD substantially in accordance with FIG. 4 A .
  • Compound 1 is in Form B characterized by an XRPD summarized in Table 2.
  • Compound 1 is in Form B characterized by a melting point of about 153.9 ⁇ 2.0° C. (onset temperature).
  • Compound 1 is in Form B characterized by a DSC substantially in accordance with FIG. 4 B .
  • Compound 1 is in Form B characterized by a TGA substantially in accordance with FIG. 4 B .
  • Also disclosed herein is a method for preparing Compound 1, such as the procedures depicted in Scheme 1.
  • the new synthetic methods and the crystallization/recrystallization procedures of Compound A via Form A or B disclosed herein overcome many issues associated with the processes reported previously, such as preparation of the key chiral intermediate with >98% optical purity, improve the purity of Compound 1 to reach the acceptance criteria in the specification, control the impurities in Compound 1 and provide many advantages over the existing processes.
  • the methods disclosed herein are especially suitable for reproducible, commercial-scale manufacture of Compound 1 in high quality and good yields.
  • BG-9 or its analogs in Scheme 1 could be asymmetrically reduced with low to excellent enantioselectivities (5% ee. to 95% ee).
  • the process of other steps are similar to those listed in Scheme 1.
  • the absolute configurations of Compound 1 was deduced to be S from the single crystal X-ray structural analysis of intermediate BG-13.
  • the solvent used in step (a) is C 1-4 alkyl alcohol (e.g., MeOH, EtOH, isopropyl alcohol), acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl 3 , dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof.
  • the solvent used in step (a) is EtOH, acetone or EtOAc.
  • the anti-solvent used in step (b) is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or H 2 O or mixtures thereof.
  • the anti-solvent used in step (b) is n-hexane, n-heptane, MTBE, or H 2 O. More preferably, the anti-solvent used in step (b) is n-hexane or n-heptane.
  • the ratio of the solvent used in step (a) and the anti-solvent used in step (b) can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected. In practice, more anti-solvent may be added to induce precipitation of the desired crystalline form.
  • the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher. Specifically, the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher, depending on whether the desired amount of crystalline form has been recovered.
  • the temperature at which step (a) is conducted is usually room temperature, although a higher temperature may be used to aid dissolution.
  • the temperature at which step (c) is conducted is 25° C. In other embodiments, the temperature at which step (c) is conducted is below 25° C., such as RT, 10° C. or 5° C.
  • the time duration for which step (c) is conducted may be adjusted by the amount of the solids which have precipitated.
  • the time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
  • the mixture of step (b) is optionally seeded with Form A to facilitate crystallization before step (c).
  • step (c) is conducted with stirring or agitation.
  • Form A is solvated.
  • the solvent used in step (a) is EtOAc, and Form A is Form A as an EtOAc solvate.
  • the resultant solvated Form A may be further converted into a different solvate by a method comprising
  • the second solvent is different from the one to dissolve Compound 1 in amorphous form, such as EtOH or acetone.
  • the second solvent is EtOH
  • the anti-solvent used in step (b) is n-heptane
  • Form A is Form A as a hetero-solvate of EtOH and n-heptane.
  • a method of preparing Form A of Compound 1 comprises suspending or slurrying Compound 1 in amorphous form in a liquid system at a temperature of or below 25° C. to precipitate Form A.
  • the liquid system is at least a solvent, or at least an anti-solvent, or a mixture of at least a solvent and at least an anti-solvent.
  • the solvent is C 1-4 alkyl alcohol (e.g., MeOH, EtOH, isopropyl alcohol), acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl 3 , dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof.
  • the solvent is EtOH, acetone or EtOAc.
  • the anti-solvent is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or H 2 O or mixtures thereof.
  • the anti-solvent is n-hexane, n-heptane, MTBE, or H 2 O. More preferably, the anti-solvent is n-hexane or n-heptane.
  • the liquid system is a mixture of at least a solvent and at least an anti-solvent.
  • the ratio of the solvent and the anti-solvent can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected.
  • the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher.
  • the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher.
  • the temperature is 25° C. In other embodiments, the temperature is conducted is below 25° C., such as RT, 10° C. or 5° C. In some embodiments, the temperature is 5° C. and the liquid system is isopropyl acetate, H 2 O, MTBE, n-heptane, NMP/H 2 O, THF/n-heptane, 1,4-Dioxane/n-heptane, EtOAc/n-heptane, or DCM/MTBE.
  • the temperature is RT
  • the liquid system is isopropyl acetate, MTBE, THF/n-heptane, 1,4-dioxane/n-heptane, EtOAc/n-heptane, or CHCl 3 /MTBE.
  • the time duration for the suspending or slurrying procedure may be adjusted by the amount of the solids which have precipitated.
  • the time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
  • the method comprises optionally adding Form A as seeds to facilitate crystallization.
  • the suspending or slurrying procedure is conducted with stirring or agitation.
  • the resultant solvated Form A may be further converted into a different solvate by suspending or slurrying the resultant solvated Form A in a different liquid system at a temperature of or below 25° C.
  • the solvent used in step (a) is acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl 3 , dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof.
  • the solvent used in step (a) is THF, CHCl 3 , acetone or EtOAc.
  • the anti-solvent used in step (b) is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or H 2 O or mixtures thereof.
  • the anti-solvent used in step (b) is n-hexane, n-heptane, MTBE, or H 2 O. More preferably, the anti-solvent used in step (b) is n-hexane or n-heptane or MTBE.
  • the ratio of the solvent used in step (a) and the anti-solvent used in step (b) can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected. In practice, more anti-solvent may be added to induce precipitation of the desired crystalline form.
  • the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher. Specifically, the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher, depending on whether the desired amount of crystalline form has been recovered.
  • the temperature at which step (a) is conducted is usually room temperature, although a higher temperature may be used to aid dissolution.
  • the temperature at which step (c) is conducted is between 50° C. to 60° C. In other embodiments, the temperature at which step (c) is conducted is 50° C. or 55° C.
  • the time duration for which step (c) is conducted may be adjusted by the amount of the solids which have precipitated.
  • the time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
  • the mixture of step (b) is optionally seeded with Form B to facilitate crystallization before step (c).
  • the resultant Form B is anhydrous non-solvated.
  • a method of preparing Form B of Compound 1 comprises suspending or slurrying Compound 1 in amorphous form or in Form A in a liquid system at a temperature of between 40° C. and 60° C. to precipitate Form B.
  • the liquid system is at least a solvent, or at least an anti-solvent, or a mixture of at least a solvent and at least an anti-solvent.
  • the solvent is acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl 3 , dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof.
  • the solvent is EtOH, acetone or EtOAc.
  • the anti-solvent is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or mixtures thereof.
  • the anti-solvent is n-hexane, n-heptane, MTBE. More preferably, the anti-solvent is n-hexane or n-heptane.
  • the liquid system is a mixture of at least a solvent and at least an anti-solvent, preferably EtOAc/n-hexane, EtOAc/n-hexane, Acetone/n-heptane, THF/n-heptane, CHCl 3 /n-heptane, CHCl 3 /MTBE, or Acetone/MTBE.
  • an anti-solvent preferably EtOAc/n-hexane, EtOAc/n-hexane, Acetone/n-heptane, THF/n-heptane, CHCl 3 /n-heptane, CHCl 3 /MTBE, or Acetone/MTBE.
  • the ratio of the solvent and the anti-solvent can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected.
  • the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher.
  • the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher.
  • the liquid is EtOAc/n-hexane (1:1), EtOAc/n-hexane (1:3), Acetone/n-heptane (1:3), THF/n-heptane (1:3), CHCl 3 /n-heptane (1:3), CHCl 3 /MTBE (1:5). or Acetone/MTBE (1:5).
  • the temperature for slurrying or suspending is between 50° C. to 60° C. In other embodiments, the temperature for slurrying or suspending is conducted is 50° C. or 55° C.
  • the time duration for the suspending or slurrying procedure may be adjusted by the amount of the solids which have precipitated.
  • the time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
  • the method comprises optionally adding Form B as seeds to facilitate crystallization.
  • the suspending or slurrying procedure is conducted with stirring or agitation.
  • a pharmaceutical composition comprises a therapeutically effective amount of Form A or B, and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is used in an oral administration.
  • the pharmaceutical composition comprises 1 wt % to 99 wt % of Form A or B.
  • the pharmaceutical composition comprises 1 wt % to 70 wt % of Form A or B.
  • the pharmaceutical composition comprises 10 wt % to 30 wt % of Form A or B.
  • Disclosed herein is a method of treating or preventing a disease associated with undesirable Btk activity in a subject by administering to a subject Form A or B.
  • Disclosed herein is a method of treating or preventing a disease selected from an allergic disease, an autoimmune disease, an inflammatory disease, a cancer, or a combination of two or more thereof in a subject by administering to the subject Form A or B.
  • a B-cell proliferative disease selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, or a combination of two or more thereof in a subject by administering to the subject Form A or B.
  • Form A or B disclosed herein can be used in manufacturing a medicament for treatment of at least one disease associated with undesirable Btk activity, in a subject.
  • Form A or B disclosed herein can be used in manufacturing a medicament for the treatment of a disease selected from an allergic disease, an autoimmune disease, an inflammatory disease, a cancer, or a combination of two or more thereof, in a subject.
  • Form A or B disclosed herein can be used in manufacturing a medicament for the treatment of a B-cell proliferative disease selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, or a combination of two or more thereof, in a subject.
  • a B-cell proliferative disease selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, or a combination of two or more thereof, in a subject.
  • the crystalline form is an approximately pure crystalline.
  • approximately pure refers to at least 85 wt %, preferably at least 95 wt %, more preferably at least 99 wt % of Form A or B disclosed herein.
  • main peaks i.e., the most characteristic, significant, unique and/or reproducible peaks
  • additional peaks may be obtained from the diffraction spectra by conventional methods.
  • the main peaks described above can be reproduced within the margin of error ( ⁇ 2 at the last given decimal place, or ⁇ 0.2 at the stated value).
  • an XRPD substantially in accordance with FIG. 2 A refers to the XRPD that show major peaks as in FIG. 2 A , wherein major peaks refer to those with the relative intensity greater than 10%, preferably greater than 20%, relative to the highest peak (with its relative intensity designated to be 100%) in FIG. 2 A .
  • solvent refers to a liquid in which Compound 1 or Form A or Form B is dissolved or partially dissolved at RT with the solubility greater than 2 mg/mL at RT, preferably greater than 10 mg/mL at RT.
  • Examples of the solvent in which Compound 1 in either amorphous form or Form A or Form B is dissolvable or partially dissolvable include, but no limited to, C 1-4 alkyl alcohol (e.g., MeOH, EtOH, isopropyl alcohol), acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl 3 , dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, acetic acid and so on.
  • C 1-4 alkyl alcohol e.g., MeOH, EtOH, isopropyl alcohol
  • acetone e.g., 4-Methyl-2-pentanone
  • EtOAc isopropyl acetate
  • anti-solvent refers to a liquid which can induce precipitation for crystallization of Form A or Form B and in which the solubility of Form A or Form B is less than 2 mg/mL at RT.
  • examples of the anti-solvent which induces precipitation for crystallization of Form A or Form B include, but not limited to, an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; H 2 O and mixtures thereof.
  • the anti-solvent for Form A or Form B is n-hexane, n-heptane, MTBE, or H 2 O.
  • terapéuticaally effective amount refers to the amount of a compound that, when administered to a subject for treating a disease, or at least one of the clinical symptoms of a disease or disorder, is sufficient to affect such treatment for the disease, disorder, or symptom.
  • the “therapeutically effective amount” can vary with the compound, the disease, disorder, and/or symptoms of the disease or disorder, severity of the disease, disorder, and/or symptoms of the disease or disorder, the age of the subject to be treated, and/or the weight of the subject to be treated. An appropriate amount in any given instance can be apparent to those skilled in the art or can be determined by routine experiments.
  • the “therapeutically effective amount” refers to the total amount of the combination objects for the effective treatment of a disease, a disorder or a condition.
  • the pharmaceutical composition comprising the compound disclosed herein can be administrated via oral, inhalation, rectal, parenteral or topical administration to a subject in need thereof.
  • the pharmaceutical composition may be a regular solid formulation such as tablets, powder, granule, capsules and the like, a liquid formulation such as water or oil suspension or other liquid formulation such as syrup, solution, suspension or the like; for parenteral administration, the pharmaceutical composition may be solution, water solution, oil suspension concentrate, lyophilized powder or the like.
  • the formulation of the pharmaceutical composition is selected from tablet, coated tablet, capsule, suppository, nasal spray or injection, more preferably tablet or capsule.
  • the pharmaceutical composition can be a single unit administration with an accurate dosage.
  • the pharmaceutical composition may further comprise additional active ingredients.
  • compositions disclosed herein can be produced by the conventional methods in the pharmaceutical field.
  • the active ingredient can be mixed with one or more excipients, then to make the desired formulation.
  • the “pharmaceutically acceptable excipient” refers to conventional pharmaceutical carriers suitable for the desired pharmaceutical formulation, for example: a diluent, a vehicle such as water, various organic solvents, etc, a filler such as starch, sucrose, etc a binder such as cellulose derivatives, alginates, gelatin and polyvinylpyrrolidone (PVP); a wetting agent such as glycerol; a disintegrating agent such as agar, calcium carbonate and sodium bicarbonate; an absorption enhancer such as quaternary ammonium compound; a surfactant such as hexadecanol; an absorption carrier such as Kaolin and soap clay; a lubricant such as talc, calcium stearate, magnesium stearate, polyethylene glycol, etc.
  • the pharmaceutical composition further comprises other pharmaceutically acceptable excipients such as a decentralized agent, a stabilizer, a thickener, a complexing agent, a buffering agent, a permeation enhancer, a polymer, aromatics, a sweetener, and a dye.
  • other pharmaceutically acceptable excipients such as a decentralized agent, a stabilizer, a thickener, a complexing agent, a buffering agent, a permeation enhancer, a polymer, aromatics, a sweetener, and a dye.
  • disease refers to any disease, discomfort, illness, symptoms or indications, and can be interchangeable with the term “disorder” or “condition”.
  • the crystalline form is characterized by X-ray powder diffraction (XRPD) pattern method, which is conducted on a PANalytical Empyrean X-ray powder diffractometer with the XRPD parameters as follows:
  • thermo-gravimetric analysis (TGA) curves are generated on a TA Q500/Q5000 TGA from TA Instruments.
  • DSC and mDSC curves are generated on a TA Q2000 DSC from TA Instruments. Detailed parameters are listed below:
  • the dynamic vapor sorption (DVS) plots are collected via a SMS (Surface Measurement Systems) DVS Intrinsic.
  • the relative humidity at 25° C. is calibrated against deliquescence point of LiCl, Mg(NO 3 ) 2 and KCl. Detailed parameters are listed below:
  • TEA 2.4 eq.
  • EtOAc 5 v
  • HOBT 1.2 eq.
  • EDCI 1.2 eq.
  • 4-phenoxybenzoic acid BG-1, 80 Kg, 1.0 eq.
  • malononitrile 1.2 eq.
  • the mixture was then stirred at RT until the reaction was completed.
  • the mixture was then centrifuged and the cake was washed with EtOAc.
  • the filtrate was washed with aqueous NaHCO 3 twice and NH 4 Cl.
  • the organic phase was washed with 1.5 N H 2 SO 4 twice and stirred.
  • the mixture was concentrated, and precipitated from methanol and purified water.
  • the reaction mixture was cooled, concentrated and then DCM was added. The final mixture was washed with saturated aqueous NH 4 Cl. The organic layer was concentrated and precipitated by charging hexane. The mixture was centrifuged and the cake was collected. The cake was dried under vacuum. This gave 82.2 Kg of the desired product.
  • ACN (5.0 v), soft water (10.0 v), KOH (5.0 eq.) was charged to a reactor and stirred for at least 15 min.
  • BG-11B (1.0 eq.) was charge to the reactor in portion-wise. The mixture was stirred until the reaction was completed. The cake was collected by centrifugation, slurried in ACN (1.0 v) and soft water (5.0 v), and dried under vacuum to give the product.
  • Step 15 Synthesis of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide (Compound 1)
  • BG-13 (6.0 Kg; 1.0 eq) was stirred in charged DCM (30 L; 5.0 v) with the presence of 5% w/w aq. NaOH (0.48 Kg; 1.2 eq.) at around 30° C. until the reaction was completed. Separated and collected organic layer. The aqueous layer was further extracted with DCM. All organic layers were combined and washed with 15% brine for twice.
  • the reaction system was concentrated under vacuum and swapped to DMF solution. Water (506.8 L; 40.0 v) was added dropwise to precipitate the solid. Centrifuged, collected the cake. The cake was dissolved in DCM and washed with 8% NaHCO 3 and 15% brine to remove DMF residue. The organic layer was concentrated and purified by silica gel (100-200 mesh column, eluted with heptane, followed by 1.2% w/w % methanol in DCM). The solution of Compound 1 was collected, concentrated and precipitate from DMF and water. The residual was centrifuged, and collected to give the crude product of Compound 1, which was confirmed to be amorphous.
  • the XRPD results found the resultant solid as a crystalline form (designated as Form A).
  • the DSC result showed an endotherm at 87.5° C. before decomposition at 274.8° C.
  • the TGA result showed a weight loss of 8.42% up to 150° C.
  • amorphous was produced ( FIG. 2 D ).
  • the 1 H NMR result showed an EtOAc content of around 9.1%, consistent with TGA weight loss.
  • the above 1 H-NMR and TGA heating confirmed Form A as an EtOAc solvated crystalline form (sometimes referred to as Form A as an EtOAc solvate) wherein the molar ratio of Compound 1 and EtOAc is about 2:1.
  • the XRPD analysis found the product obtained from crystallization from EtOH/n-heptane and the product obtained from crystallization from acetone/n-heptane are crystalline; see FIG. 3 A .
  • FIG. 3 A shows that the XRPD patterns of Form A obtained from 2:1 EtOAc/n-heptane (v/v) in Example 2, Form A obtained from EtOH/n-heptane and Form A obtained from acetone/n-heptane comprise the diffraction peaks of substantially the same positions, although their relative intensities vary.
  • Form A with increased crystallinity was prepared by slurry of amorphous in EtOH/n-heptane (1:3, v/v) at RT, as shown in FIG. 3 B (also confirmed the crystalline structure of Form A, referred to as Form A obtained from slurry conversion of EtOH/n-heptane).
  • the TGA curve of Form A obtained from slurry conversion of EtOH/n-heptane in FIG. 3 C showed a two-step loss of 6.8% up to 150° C.
  • the 1 H-NMR result of Form A obtained from slurry conversion of EtOH/n-heptane in FIG. 3 D showed an EtOH content of 0.4% and n-heptane content of 4.4% without EtOAc residual. After desolvation via TGA heating, amorphous was also produced as shown in FIG. 3 E .
  • the resultant solid was then subject to various characterizations including XRPD ( FIG. 4 A ), TGA/DSC ( FIG. 4 B ), 1 H NMR ( FIG. 4 C ) and 13 C-NMR (DMSO-d 6 ) ( FIG. 4 D ).
  • the XRPD result found the resultant solid as a crystalline form different from the above Form A (designated as Form B).
  • the DSC result showed a neat melting endotherm at 153.9° C. (onset temperature).
  • the TGA result showed a two-step weight loss of 2.3% up to 140° C., indicating Form B is anhydrous non-solvated.
  • the 1 H-NMR showed no solvent residual EtOAc that appeared in Form A, which is consistent with the TGA result.
  • Example 2 250 mg of Compound 1 in amorphous form prepared in Example 1 was added into a 20-mL glass vial. 1.0 mL of THF was added into the vial and stirred at RT until all solids were dissolved. 1.0 mL of n-heptane was added to induce precipitate. The mixture was equilibrated at RT for 1 hr. About 4 mg of Form B of Compound 1 prepared in the above Example 4 was added as seed and the mixture was stirred at RT for 1.5 hrs. Additional 2.0 mL of n-heptane was added to induce more solids. About 4 mg of Form B of Compound 1 prepared in the above Example 4 was added again as seed and the mixture was stirred at 50° C. for 22 hrs. The cake was collected by vacuum filter and dried at 50° C. for 16 hrs to obtain the solid (168.2 mg, yield of ⁇ 72%).
  • Example 5A and 5B are Form B as their XRPD patterns ( FIG. 4 G ) are consistent with that of Example 4 ( FIG. 4 A ).
  • Example 6 Slurry Conversion from Compound 1 of Form A or Amorphous Form at Different Temperatures
  • Example 6A Slurry Conversion from Form A at 5° C.
  • Example 6B Slurry Conversion from Form A at RT
  • Form B D10 EtOAc/n-heptane (3:1) Form B D11 EtOAc/n-heptane (1:1) Form B D12 EtOAc/n-heptane (1:3) Form B *Small part of Form A was observed, indicating Form B is thermodynamically more stable under this condition.
  • Form A and Form B were exposed under EtOAc/n-heptane system with a series of solvent ratios and temperatures (10° C., 20 ⁇ 2° C., and 50° C.), which indicated that Form B is thermodynamically more stable than Form A at 50° C., and at room temperature with a volume fraction of EtOAc decreased to 0.25.
  • anhydrous non-solvated Form B tends to be the stable crystalline form under conditions with elevated temperature, decreased EtOAc content, seed loading and extended equilibration time.
  • BG-13 was confirmed to be a (2R, 3R)-dibenzoyl tartaric acid (L-DBTA) salt and the molar ratio of freebase to L-DBTA is 2:1.
  • Configuration of both carbons (C32 and C32′) in L-DBTA was confirmed to be R.
  • Configuration of C6 in freebase was determined to be S, as shown in FIG. 6 A to FIG. 6 C .
  • a powder X-ray diffraction pattern method was also used to characterize the structure of the single crystals, as shown in FIG. 6 D .
  • Form B of Compound 1 was tested for inhibition of BTK kinase (aa2-659, Carna Biosciences) in assays based on the time-resolved fluorescence-resonance energy transfer (TR-FRET) methodology.
  • the assays were carried out in 384-well low volume black plates in a reaction mixture containing BTK kinase, 5 ⁇ M ATP, 2 ⁇ M peptide substrate and 0-10 ⁇ M Form B of Compound 1 of Example 4 in buffer containing 50 mM Tris pH7.4, 10 mM MgCl2, 2 mM MnCl 2 , 0.1 mM EDTA, 1 mM DTT, 0.005% Tween-20, 20 nM SEB and 0.01% BSA.
  • the kinase was incubated with compound for 60 minutes at room temperature and the reaction was initiated by the addition of ATP and peptide substrate. After reaction at room temperature for 60 minutes, an equal volume of stop/detection solution was added according to the manufacture's instruction (CisBio Bioassays).
  • the stop/detection solution contained Eu 3+ cryptate-conjugated mouse monoclonal antibody (PT66) anti-phosphotyrosine and XL665-conjugated streptavidin in buffer containing 50 mM HEPES pH 7.0, 800 mM KF, 20 mM EDTA, and 0.1% BSA.
  • TR-FRET signals ratio of fluorescence emission at 665 nm over emission at 620 nm with excitation at 337 nm wavelength
  • TR-FRET signals ratio of fluorescence emission at 665 nm over emission at 620 nm with excitation at 337 nm wavelength
  • Phosphorylation of peptide substrate led to the binding of anti-phosphotyrosine antibody to the biotinylated peptide substrate, which places fluorescent donor (Eu 3+ crypate) in close proximity to the accepter (Streptavidin-XL665), thus resulting in a high degree of fluorescence resonance energy transfer from the donor fluorophore (at 620 nm) to the acceptor fluorophore (at 665 nm).
  • Inhibition of BTK kinase activity resulted in decrease of the TR-FRET signal.
  • the IC 50 for Compound 1 in Form B was derived from fitting the data to the four-parameter logistic equation by Graphpad Prism software.
  • the IC50 of Compound 1 in Form B is 1 nM.
  • Form B was profiled against a panel of 367 kinases at 2 ⁇ M at Reaction Biology Corp.
  • Form B displayed less than 70% inhibition against 358 kinases, and greater than 70% inhibition against 9 kinases including BTK.

Abstract

The present invention relates to a crystalline form of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide (Compound 1) for inhibiting Btk, methods of preparation thereof and pharmaceutical compositions, and use of the crystalline form above in the treatment of a disease, or in the manufacturing of a medicament for the treatment of a disease, such as an allergic disease, an autoimmune disease, an inflammatory disease, and a cancer.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a U.S. National Stage Application under 35 U.S.C. § 371 of International Application No. PCT/CN2018/074108, filed Jan. 25, 2018, which claims the benefit of International Patent Application No. PCT/CN2017/072553 filed Jan. 25, 2017, the disclosures of which are hereby incorporated by reference in their entireties for all purposes.
FIELD OF THE INVENTION
Disclosed herein are crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide (hereinafter Compound 1); methods for preparing crystalline forms of Compound 1; pharmaceutical compositions comprising crystalline forms of Compound 1 and a pharmaceutically acceptable carrier; and methods of using crystalline forms of Compound 1 as a Btk inhibitor for treating or preventing diseases.
BACKGROUND OF THE INVENTION
Bruton's tyrosine kinase (Btk) belongs to the Tec tyrosine kinase family (Vetrie et al., Nature 361: 226-233, 1993; Bradshaw, Cell Signal. 22: 1175-84, 2010). Btk is primarily expressed in most hematopoietic cells such as B cells, mast cells and macrophages (Smith et al., J. Immunol. 152: 557-565, 1994) and is localized in bone marrow, spleen and lymph node tissue. Btk plays important roles in B-cell receptor (BCR) and FcR signaling pathways, which involve in B-cell development, differentiation (Khan, Immunol. Res. 23: 147, 2001). Btk is activated by upstream Src-family kinases. Once activated, Btk in turn phosphorylates PLC gamma, leading to effects on B-cell function and survival (Humphries et al., J Biol. Chem. 279: 37651, 2004).
These signaling pathways must be precisely regulated. Mutations in the gene encoding Btk cause an inherited B-cell specific immunodeficiency disease in humans, known as X-linked a gamma-globulinemia (XLA) (Conley et al., Annu. Rev. Immunol. 27: 199-227, 2009). Aberrant BCR-mediated signaling may result in dysregulated B-cell activation leading to a number of autoimmune and inflammatory diseases. Preclinical studies show that Btk deficient mice are resistant to developing collagen-induced arthritis. Moreover, clinical studies of Rituxan, a CD20 antibody to deplete mature B-cells, reveal the key role of B-cells in a number of inflammatory diseases such as rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis (Gurcan et al., Int. Immunopharmacol. 9: 10-25, 2009). Therefore, Btk inhibitors can be used to treat autoimmune and/or inflammatory diseases.
In addition, aberrant activation of Btk plays an important role in pathogenesis of B-cell lymphomas indicating that inhibition of Btk is useful in the treatment of hematological malignancies (Davis et al., Nature 463: 88-92, 2010). Preliminary clinical trial results showed that the Btk inhibitor PCI-32765 was effective in treatment of several types of B-cell lymphoma (for example, 54th American Society of Hematology (ASH) annual meeting abstract, December 2012: 686 The Bruton's Tyrosine Kinase (Btk) Inhibitor, Ibrutinib (PCI-32765), Has Preferential Activity in the ABC Subtype of Relapsed/Refractory De Novo Diffuse Large B-Cell Lymphoma (DLBCL): Interim Results of a Multicenter, Open-Label, Phase! Study). Because Btk plays a central role as a mediator in multiple signal transduction pathways, inhibitors of Btk are of great interest as anti-inflammatory and/or anti-cancer agents (Mohamed et al., Immunol. Rev. 228: 58-73, 2009; Pan, Drug News perspect 21: 357-362, 2008; Rokosz et al., Expert Opin. Ther. Targets 12: 883-903, 2008; Uckun et al., Anti-cancer Agents Med. Chem. 7: 624-632, 2007; Lou et al, J. Med. Chem. 55(10): 4539-4550, 2012).
International application WO2014173289A disclosed a series of fused heterocyclic compounds as Btk inhibitors. In particular, WO2014173289A disclosed (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide
Figure US11555038-20230117-C00002
Compound 1 is a potent, specific and irreversible BTK kinase inhibitor. The data generated in preclinical studies using biochemical, cell based and animal studies have suggested that Compound 1 is of significant benefit in inhibiting tumor growth in B-cell malignancies. Compound 1 was shown to be more selective than ibrutinib for inhibition of BTK vs. EGFR, FGR, FRK, HER2, HER4, ITK, JAK3, LCK, and TEC, indicating that Compound 1 will have less side-effects than ibrutinib in clinic.
However, Compound 1 prepared in accordance with WO2014173289A was found to be in amorphous form, as confirmed by the X-Ray Powder Diffraction (hereinafter XRPD) pattern of FIG. 1A (the existence of a large bump distributed in a wide range) and the TGA/mDSC curves of FIG. 1B (no substantial glass transition signal). The amorphous form of Compound 1 presents some challenges for drug formulation due to its low stability and difficulties to handle. Therefore, there is a need to develop new forms of Compound 1, especially crystalline forms, which are stable and easy to handle and to process in the manufacture and preparation of drug formulations.
SUMMARY OF THE INVENTION
The inventors of the present invention have found solvated or non-solvated crystalline forms of Compound 1, which are stable and easy to handle and to process in the manufacture and preparation of drug formulations compared with the amorphous form of Compound 1. Particularly, the inventors found an anhydrous non-solvated crystalline form (i.e., Form B herein) is suitable for drug formulation due to its high melting point, non-hygroscopicity and good mechanical stability.
In a first aspect, disclosed herein is a crystalline form of Compound 1,
Figure US11555038-20230117-C00003
In some embodiments, the crystalline form of Compound 1 is solvated (herein referred to as “Form A” or “Form A as a solvate” or “solvated crystalline form”).
In some embodiments, the crystalline form of Compound 1 is anhydrous non-solvated (herein referred to as “Form B” or “non-solvated crystalline form”).
In a second aspect, disclosed herein is a method of preparing Compound 1.
In a third aspect, disclosed herein is a method of preparing Form A disclosed herein.
In a forth aspect, disclosed herein is a method of preparing Form B disclosed herein.
In a fifth aspect, disclosed herein is a pharmaceutical composition comprising a therapeutically effective amount of Form A or B disclosed herein and a pharmaceutically acceptable carrier.
In a sixth aspect, disclosed herein is a method for treating a disease associated with undesirable Btk activity in a subject by administering to the subject Form A or B disclosed herein. Preferably, the disease is an allergic disease, an autoimmune disease, an inflammatory disease, or a cancer, or a combination of two or more thereof. More preferably, the disease is a B-cell proliferative disease, selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, or chronic lymphocytic leukemia, or a combination of two or more thereof. In some embodiment, the inflammatory disease is selected from rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis.
In a seventh aspect, disclosed herein is a use of Form A or B disclosed herein in manufacturing a medicament for treatment of at least one disease associated with undesirable Btk activity. Preferably, the disease is an allergic disease, an autoimmune disease, an inflammatory disease, or a cancer, or a combination of two or more thereof. More preferably, the disease is a B-cell proliferative disease, selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, or chronic lymphocytic leukemia, or a combination of two or more thereof. In some embodiment, the inflammatory disease is selected from rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis.
BRIEF DESCRIPTIONS OF THE DRAWINGS
FIG. 1A shows the XRPD pattern of the amorphous form of Compound 1.
FIG. 1B shows the TGA/mDSC curves of amorphous form of Compound 1.
FIG. 2A shows the XRPD pattern of Form A (crystallization from EtOAc/n-heptane).
FIG. 2B shows the TGA/DSC curves of Form A (crystallization from EtOAc/n-heptane).
FIG. 2C shows the 1H-NMR of Form A (crystallization from EtOAc/n-heptane).
FIG. 2D shows XRPD overlay of Form A (crystallization from EtOAc/n-heptane) before and after heating.
FIG. 3A shows the XRPD pattern of Form A (crystallization from EtOH/n-heptane) and Form A (crystallization from acetone/n-heptane).
FIG. 3B shows the XRPD pattern of Form A obtained from slurry conversion of EtOH/n-heptane.
FIG. 3C shows the TGA curve of Form A obtained from slurry conversion of EtOH/n-heptane.
FIG. 3D shows the 1H-NMR of Form A obtained from slurry conversion of EtOH/n-heptane.
FIG. 3E shows the XRPD overlay of Form A obtained from slurry conversion of EtOH/n-heptane before and after heating.
FIG. 4A shows the XRPD pattern of Crystalline Form B.
FIG. 4B shows the TGA/DSC curves of Form B.
FIG. 4C shows the 1H-NMR of Crystalline Form B.
FIG. 4D shows the 13C-NMR of Crystalline Form B.
FIG. 4E shows DVS plot of Crystalline Form B.
FIG. 4F shows XRPD patterns of Form B pre and post DVS.
FIG. 4G shows XRPD overlay of Form B of Example 4, Example 5A and Example 5B.
FIG. 4H shows XRPD overlay of Form B before and after grinding.
FIG. 5A shows XRPD overlay for slurry at RT starting from amorphous Compound 1.
FIG. 5B shows XRPD overlay for slurry at 50° C. starting from amorphous Compound 1.
FIG. 5C shows XRPD overlay for competitive slurry of Form A and B at 10° C.
FIG. 5D shows XRPD overlay for competitive slurry of Form A and B at RT.
FIG. 5E shows XRPD overlay for competitive slurry of Form A and B at 50° C.
FIG. 6A shows the absolute structure of single crystal of BG-13.
FIG. 6B illustrates hydrogen bonds of single crystal of BG-13.
FIG. 6C shows a crystal packing of single crystal of BG-13.
FIG. 6D shows the XRPD pattern of single crystal of BG-13.
DETAILED DESCRIPTION OF THE INVENTION
The inventors have found that Compound 1 can exist in crystalline forms, either solvated or non-solvated.
In some embodiments, Compound 1 is in solvated crystalline form (i.e., Form A). In a preferred embodiment, Form A is solvated with EtOAc (also referred to as “Form A as an EtOAc solvate”). In other embodiments, Form A is solvated with other solvent or anti-solvent or is to form a hetero-solvate.
In some embodiments, Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 5.3±0.2°, 16.9±0.2°, 18.6±0.2°, 19.8±0.2° and 20.6±0.2°.
In some embodiments, Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 5.3±0.2°, 16.9±0.2°, 18.6±0.2°, 19.8±0.2° and 20.6±0.2°.
In some embodiments, Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 5.3±0.2°, 10.9±0.2°, 16.9±0.2°, 18.6±0.2°, 19.8±0.2°, 20.6±0.2°, 21.1±0.2°, 22.5±0.2° and 22.8±0.20.
In some embodiments, Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 5.3±0.2°, 10.9±0.2°, 11.2±0.2°, 13.4±0.2°, 14.4±0.2°, 16.9±0.2°, 18.6±0.2°, 19.8±0.2°, 20.6±0.2°, 21.1±0.2°, 21.7±0.2°, 22.5±0.2°, 22.8±0.2°, 23.6±0.2° and 24.3±0.2°.
In some embodiments, Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 5.3±0.2°, 10.9±0.2°, 11.2±0.2°, 12.0±0.2°, 13.4±0.2°, 14.1±0.2°, 14.4±0.2°, 16.9±0.2°, 18.1±0.2°, 18.6±0.2°, 19.8±0.2°, 20.6±0.2°, 21.1±0.2°, 21.7±0.2°, 22.5±0.2°, 22.8±0.2°, 23.6±0.2°, 24.0±0.2° and 24.3±0.2°.
In some embodiments, Compound 1 is in Form A characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 5.3±0.2°, 9.2±0.2°, 10.9±0.2°, 11.2±0.2°, 12.0±0.2°, 13.4±0.2°, 14.1±0.2°, 14.4±0.2°, 15.5±0.2°, 16.9±0.2°, 17.7±0.2°, 18.1±0.2°, 18.6±0.2°, 19.8±0.2°, 20.6±0.2°, 21.1±0.2°, 21.7±0.2°, 22.5±0.2°, 22.8±0.2°, 23.6±0.2°, 24.0±0.2°, 24.3±0.2° and 26.4±0.2°.
In some embodiments, Compound 1 is in Form A characterized by an XRPD substantially in accordance with FIG. 2A.
In some embodiments, Compound 1 is in Form A characterized by an XRPD summarized in Table 1.
TABLE 1
X-ray Diffraction Pattern of Form A of Compound 1
Diffraction Relative
Peak# angle (2 theta) Spacing intensity
1 5.257 16.80987 75.65
2 6.847 12.91043 9.90
3 8.868 9.97248 12.65
4 9.217 9.59468 16.83
5 10.947 8.08267 54.46
6 11.228 7.88084 38.01
7 11.972 7.39264 19.97
8 13.383 6.61638 46.72
9 14.131 6.26740 34.23
10 14.449 6.13029 39.26
11 15.532 5.70515 18.97
12 16.900 5.24643 87.03
13 17.754 4.99588 23.00
14 18.126 4.89410 29.17
15 18.605 4.76923 100.00
16 19.842 4.47464 88.29
17 20.560 4.31994 90.99
18 21.058 4.21899 57.80
19 21.782 4.08036 41.63
20 22.453 3.95982 74.39
21 22.822 3.89668 67.54
22 23.607 3.76878 37.10
23 24.061 3.69869 33.71
24 24.323 3.65943 36.77
25 25.306 3.51949 35.61
26 26.400 3.37615 16.09
27 27.115 3.28872 13.51
28 28.339 3.14940 7.03
29 29.252 3.05308 8.39
30 30.048 2.97402 8.02
31 30.670 2.91511 6.82
32 35.033 2.56145 3.45
33 35.860 2.50421 3.48
In some preferred embodiments, Compound 1 is in Form A as an EtOAc solvate characterized by a DSC substantially in accordance with FIG. 2B.
In some preferred embodiments, Compound 1 is in Form A as an EtOAc solvate characterized by a TGA substantially in accordance with FIG. 2B.
In some embodiments, Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 15.1±0.2°, 20.7±0.2° and 22.8±0.2°.
In some embodiments, Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 15.1±0.2°, 15.5±0.2°, 19.9±0.2°, 20.7±0.2°, 21.7±0.2° and 22.8±0.2°.
In some embodiments, Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 15.1±0.2°, 15.5±0.2°, 17.0±0.2°, 19.9±0.2°, 20.0±0.2°, 20.7±0.2°, 21.7±0.2°, 22.8±0.2° and 24.2±0.20.
In some embodiments, Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 6.3±0 0.2°, 9.5±0.2°, 15.1±0.2°, 15.5±0.2°, 16.5±0.2°, 17.0±0.2°, 19.9±0.2°, 20.0±0.2°, 20.7±0.2°, 21.7±0.2°, 22.8±0.2°, 24.2±0.2° and 25.1±0.2°.
In some embodiments, Compound 1 is in Form B characterized by an XRPD comprising diffraction peaks having 2θ angle values independently selected from: approximately 6.3±0 0.2°, 9.5±0.2°, 12.4±0.2°, 13.2±0.2°, 15.1±0.2°, 15.5±0.2°, 16.5±0.2°, 17.0±0.2°, 18.0±0.2°, 18.9±0.2°, 19.9±0.2°, 20.0±0.2°, 20.7±0.2°, 21.7±0.2°, 22.8±0.2°, 24.2±0.2°, 25.1±0.2°, 25.8±0.2°, 26.7±0.2° and 27.6±0.2°.
In some embodiments, Compound 1 is in Form B characterized by an XRPD substantially in accordance with FIG. 4A.
I In some embodiments, Compound 1 is in Form B characterized by an XRPD summarized in Table 2.
TABLE 2
X-ray Diffraction Pattern of Compound 1 Crystalline Form B
Diffraction Relative
Peak# angle (2 theta) Spacing intensity
1 6.266 14.10502 21.70
2 9.483 9.32619 22.34
3 11.455 7.72521 3.92
4 12.395 7.14115 11.38
5 12.832 6.89900 12.73
6 13.252 6.68146 12.21
7 15.095 5.86950 100.00
8 15.513 5.71236 66.31
9 16.530 5.36287 21.11
10 17.016 5.21086 44.01
11 18.032 4.91940 22.41
12 18.839 4.71056 22.97
13 18.946 4.68427 26.61
14 19.519 4.54788 9.92
15 19.897 4.46247 61.31
16 20.081 4.42199 31.97
17 20.687 4.29383 97.71
18 21.722 4.09137 58.94
19 22.800 3.90041 97.94
20 24.243 3.67138 32.46
21 24.822 3.58706 13.83
22 25.110 3.54655 27.64
23 25.757 3.45886 12.94
24 26.306 3.38790 11.20
25 26.732 3.33487 10.85
26 27.334 3.26277 10.51
27 27.665 3.22453 13.80
28 28.480 3.13406 4.80
29 29.298 3.04844 5.98
30 29.765 3.00164 8.35
31 30.128 2.96635 5.63
32 30.789 2.90412 5.73
33 31.326 2.85555 6.85
34 31.613 2.83028 6.13
35 32.049 2.79277 8.88
36 32.983 2.71581 5.91
37 34.973 2.56565 8.57
38 37.038 2.42722 1.32
39 38.049 2.36504 3.63
In some preferred embodiments, Compound 1 is in Form B characterized by a melting point of about 153.9±2.0° C. (onset temperature).
In some preferred embodiments, Compound 1 is in Form B characterized by a DSC substantially in accordance with FIG. 4B.
In some preferred embodiments, Compound 1 is in Form B characterized by a TGA substantially in accordance with FIG. 4B.
Also disclosed herein is a method for preparing Compound 1, such as the procedures depicted in Scheme 1. The new synthetic methods and the crystallization/recrystallization procedures of Compound A via Form A or B disclosed herein overcome many issues associated with the processes reported previously, such as preparation of the key chiral intermediate with >98% optical purity, improve the purity of Compound 1 to reach the acceptance criteria in the specification, control the impurities in Compound 1 and provide many advantages over the existing processes. Notably, the methods disclosed herein are especially suitable for reproducible, commercial-scale manufacture of Compound 1 in high quality and good yields. In an alternative process, BG-9 or its analogs in Scheme 1 could be asymmetrically reduced with low to excellent enantioselectivities (5% ee. to 95% ee). The process of other steps are similar to those listed in Scheme 1. And the absolute configurations of Compound 1 was deduced to be S from the single crystal X-ray structural analysis of intermediate BG-13.
Figure US11555038-20230117-C00004
Also disclosed herein are methods of preparing Form A or Form B.
Disclosed herein is a method of preparing Form A of Compound 1, comprising:
    • (a) Dissolving Compound 1 in amorphous form in a solvent;
    • (b) Adding an anti-solvent into the solution of step (a) to induce precipitation;
    • (c) Keeping the mixture of step (b) at a temperature of or below 25° C. to precipitate Form A.
In some embodiments, the solvent used in step (a) is C1-4alkyl alcohol (e.g., MeOH, EtOH, isopropyl alcohol), acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl3, dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof. Preferably, the solvent used in step (a) is EtOH, acetone or EtOAc.
In some embodiments, the anti-solvent used in step (b) is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or H2O or mixtures thereof. Preferably, the anti-solvent used in step (b) is n-hexane, n-heptane, MTBE, or H2O. More preferably, the anti-solvent used in step (b) is n-hexane or n-heptane.
The ratio of the solvent used in step (a) and the anti-solvent used in step (b) can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected. In practice, more anti-solvent may be added to induce precipitation of the desired crystalline form. In some embodiments, the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher. Specifically, the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher, depending on whether the desired amount of crystalline form has been recovered.
The temperature at which step (a) is conducted is usually room temperature, although a higher temperature may be used to aid dissolution.
In some embodiments, the temperature at which step (c) is conducted is 25° C. In other embodiments, the temperature at which step (c) is conducted is below 25° C., such as RT, 10° C. or 5° C.
The time duration for which step (c) is conducted may be adjusted by the amount of the solids which have precipitated. The time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
In some embodiments, the mixture of step (b) is optionally seeded with Form A to facilitate crystallization before step (c).
In some embodiment, step (c) is conducted with stirring or agitation.
In some embodiments, Form A is solvated. In some embodiment, the solvent used in step (a) is EtOAc, and Form A is Form A as an EtOAc solvate.
In some embodiments, the resultant solvated Form A may be further converted into a different solvate by a method comprising
    • (a′) Dissolving the resultant Form A in a second solvent;
    • (b′) Adding a second anti-solvent into the solution of step (a′) to induce precipitation;
    • (c′) Keeping the mixture of step (b′) at a temperature of or below 25° C. to precipitate Form A as a different solvate.
In some embodiment, the second solvent is different from the one to dissolve Compound 1 in amorphous form, such as EtOH or acetone. In a further embodiment, the second solvent is EtOH, the anti-solvent used in step (b) is n-heptane, and Form A is Form A as a hetero-solvate of EtOH and n-heptane.
Alternatively, a method of preparing Form A of Compound 1 comprises suspending or slurrying Compound 1 in amorphous form in a liquid system at a temperature of or below 25° C. to precipitate Form A.
In some embodiments, the liquid system is at least a solvent, or at least an anti-solvent, or a mixture of at least a solvent and at least an anti-solvent. The solvent is C1-4alkyl alcohol (e.g., MeOH, EtOH, isopropyl alcohol), acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl3, dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof. Preferably, the solvent is EtOH, acetone or EtOAc. The anti-solvent is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or H2O or mixtures thereof. Preferably, the anti-solvent is n-hexane, n-heptane, MTBE, or H2O. More preferably, the anti-solvent is n-hexane or n-heptane.
Preferably, the liquid system is a mixture of at least a solvent and at least an anti-solvent. The ratio of the solvent and the anti-solvent can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected. In some embodiments, the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher. Specifically, the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher.
In some embodiments, the temperature is 25° C. In other embodiments, the temperature is conducted is below 25° C., such as RT, 10° C. or 5° C. In some embodiments, the temperature is 5° C. and the liquid system is isopropyl acetate, H2O, MTBE, n-heptane, NMP/H2O, THF/n-heptane, 1,4-Dioxane/n-heptane, EtOAc/n-heptane, or DCM/MTBE. In other embodiments, the temperature is RT, and the liquid system is isopropyl acetate, MTBE, THF/n-heptane, 1,4-dioxane/n-heptane, EtOAc/n-heptane, or CHCl3/MTBE.
The time duration for the suspending or slurrying procedure may be adjusted by the amount of the solids which have precipitated. The time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
In some embodiments, the method comprises optionally adding Form A as seeds to facilitate crystallization.
In some embodiment, the suspending or slurrying procedure is conducted with stirring or agitation.
In some embodiments, the resultant solvated Form A may be further converted into a different solvate by suspending or slurrying the resultant solvated Form A in a different liquid system at a temperature of or below 25° C.
Also disclosed herein is a method of preparing Form B of Compound 1, comprising:
    • (a) Dissolving Compound 1 in Form A or in amorphous form in a solvent;
    • (b) Adding an anti-solvent into the solution of step (a) to induce precipitation;
    • (c) Keeping the mixture of step (b) at a temperature of between 40° C. and 60° C. to precipitate Form B.
In some embodiments, the solvent used in step (a) is acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl3, dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof. Preferably, the solvent used in step (a) is THF, CHCl3, acetone or EtOAc.
In some embodiments, the anti-solvent used in step (b) is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or H2O or mixtures thereof. Preferably, the anti-solvent used in step (b) is n-hexane, n-heptane, MTBE, or H2O. More preferably, the anti-solvent used in step (b) is n-hexane or n-heptane or MTBE.
The ratio of the solvent used in step (a) and the anti-solvent used in step (b) can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected. In practice, more anti-solvent may be added to induce precipitation of the desired crystalline form. In some embodiments, the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher. Specifically, the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher, depending on whether the desired amount of crystalline form has been recovered.
The temperature at which step (a) is conducted is usually room temperature, although a higher temperature may be used to aid dissolution.
In some embodiments, the temperature at which step (c) is conducted is between 50° C. to 60° C. In other embodiments, the temperature at which step (c) is conducted is 50° C. or 55° C.
The time duration for which step (c) is conducted may be adjusted by the amount of the solids which have precipitated. The time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
In some embodiments, the mixture of step (b) is optionally seeded with Form B to facilitate crystallization before step (c).
In some embodiments, the resultant Form B is anhydrous non-solvated.
Alternatively, a method of preparing Form B of Compound 1 comprises suspending or slurrying Compound 1 in amorphous form or in Form A in a liquid system at a temperature of between 40° C. and 60° C. to precipitate Form B.
In some embodiments, the liquid system is at least a solvent, or at least an anti-solvent, or a mixture of at least a solvent and at least an anti-solvent. The solvent is acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl3, dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, or acetic acid or mixtures thereof. Preferably, the solvent is EtOH, acetone or EtOAc. The anti-solvent is an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; or mixtures thereof. Preferably, the anti-solvent is n-hexane, n-heptane, MTBE. More preferably, the anti-solvent is n-hexane or n-heptane.
Preferably, the liquid system is a mixture of at least a solvent and at least an anti-solvent, preferably EtOAc/n-hexane, EtOAc/n-hexane, Acetone/n-heptane, THF/n-heptane, CHCl3/n-heptane, CHCl3/MTBE, or Acetone/MTBE.
The ratio of the solvent and the anti-solvent can be easily determined by a skilled person in the art once the solvent and the anti-solvent have been selected. In some embodiments, the volumetric ratio of the solvent/anti-solvent ratio is from 2:1 to 1:7 or higher. Specifically, the volumetric ratio of the solvent:anti-solvent ratio is 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or higher. Preferably, the liquid is EtOAc/n-hexane (1:1), EtOAc/n-hexane (1:3), Acetone/n-heptane (1:3), THF/n-heptane (1:3), CHCl3/n-heptane (1:3), CHCl3/MTBE (1:5). or Acetone/MTBE (1:5).
In some embodiments, the temperature for slurrying or suspending is between 50° C. to 60° C. In other embodiments, the temperature for slurrying or suspending is conducted is 50° C. or 55° C.
The time duration for the suspending or slurrying procedure may be adjusted by the amount of the solids which have precipitated. The time duration may be 1 hours, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours or longer.
In some embodiments, the method comprises optionally adding Form B as seeds to facilitate crystallization.
In some embodiment, the suspending or slurrying procedure is conducted with stirring or agitation.
Also disclosed herein is a pharmaceutical composition comprises a therapeutically effective amount of Form A or B, and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition is used in an oral administration. In some preferred embodiments, the pharmaceutical composition comprises 1 wt % to 99 wt % of Form A or B. In some more preferred embodiments, the pharmaceutical composition comprises 1 wt % to 70 wt % of Form A or B. In some most embodiments, the pharmaceutical composition comprises 10 wt % to 30 wt % of Form A or B.
Disclosed herein is a method of treating or preventing a disease associated with undesirable Btk activity in a subject by administering to a subject Form A or B.
Disclosed herein is a method of treating or preventing a disease selected from an allergic disease, an autoimmune disease, an inflammatory disease, a cancer, or a combination of two or more thereof in a subject by administering to the subject Form A or B.
Disclosed herein is a method of treating or preventing a B-cell proliferative disease, selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, or a combination of two or more thereof in a subject by administering to the subject Form A or B.
Form A or B disclosed herein can be used in manufacturing a medicament for treatment of at least one disease associated with undesirable Btk activity, in a subject.
Form A or B disclosed herein can be used in manufacturing a medicament for the treatment of a disease selected from an allergic disease, an autoimmune disease, an inflammatory disease, a cancer, or a combination of two or more thereof, in a subject.
Form A or B disclosed herein can be used in manufacturing a medicament for the treatment of a B-cell proliferative disease selected from chronic lymphocytic, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, mantle cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, or a combination of two or more thereof, in a subject.
Definitions
Unless specifically defined elsewhere in this document, all other technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which this invention belongs.
As used herein, including the appended claims, the singular forms of words such as “a”, “an”, and “the”, include their corresponding plural references unless the context clearly dictates otherwise. Thus, for example, reference to “a crystalline form” includes one or more of such different crystalline forms and reference to “the method” includes reference to equivalent steps and methods know to those of ordinary skill in the art that could be modified or substituted for the methods described herein.
As disclosed herein, the crystalline form is an approximately pure crystalline. The term “approximately pure” as herein used refers to at least 85 wt %, preferably at least 95 wt %, more preferably at least 99 wt % of Form A or B disclosed herein.
For crystalline forms disclosed herein, only the main peaks (i.e., the most characteristic, significant, unique and/or reproducible peaks) are summarized; additional peaks may be obtained from the diffraction spectra by conventional methods. The main peaks described above can be reproduced within the margin of error (±2 at the last given decimal place, or ±0.2 at the stated value).
As disclosed herein, “an XRPD substantially in accordance with FIG. 2A” refers to the XRPD that show major peaks as in FIG. 2A, wherein major peaks refer to those with the relative intensity greater than 10%, preferably greater than 20%, relative to the highest peak (with its relative intensity designated to be 100%) in FIG. 2A.
The term “solvent” used herein refers to a liquid in which Compound 1 or Form A or Form B is dissolved or partially dissolved at RT with the solubility greater than 2 mg/mL at RT, preferably greater than 10 mg/mL at RT. Examples of the solvent in which Compound 1 in either amorphous form or Form A or Form B is dissolvable or partially dissolvable include, but no limited to, C1-4alkyl alcohol (e.g., MeOH, EtOH, isopropyl alcohol), acetone, 4-Methyl-2-pentanone, EtOAc, isopropyl acetate, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane, acetonitrile, CHCl3, dichloromethane, toluene, N,N-dimethylacetamide, dimethylsulfoxide, 1-methyl-2-pyrrolidone, acetic acid and so on.
The term “anti-solvent” used herein refers to a liquid which can induce precipitation for crystallization of Form A or Form B and in which the solubility of Form A or Form B is less than 2 mg/mL at RT. Examples of the anti-solvent which induces precipitation for crystallization of Form A or Form B include, but not limited to, an aliphatic hydrocarbon, which preferably is a hexane such as n-hexane, a heptane such as n-heptane, a cyclohexane and/or petroleum ether; and an aliphatic ether, which preferably is MTBE; H2O and mixtures thereof. Preferably, the anti-solvent for Form A or Form B is n-hexane, n-heptane, MTBE, or H2O.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word “comprise”, and variations such as “comprises” and “comprising”, will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integer or step. When used herein the term “comprising” can be substituted with the term “containing” or sometimes when used herein with the term “having”.
The term “therapeutically effective amount” as herein used, refers to the amount of a compound that, when administered to a subject for treating a disease, or at least one of the clinical symptoms of a disease or disorder, is sufficient to affect such treatment for the disease, disorder, or symptom. The “therapeutically effective amount” can vary with the compound, the disease, disorder, and/or symptoms of the disease or disorder, severity of the disease, disorder, and/or symptoms of the disease or disorder, the age of the subject to be treated, and/or the weight of the subject to be treated. An appropriate amount in any given instance can be apparent to those skilled in the art or can be determined by routine experiments. In the case of combination therapy, the “therapeutically effective amount” refers to the total amount of the combination objects for the effective treatment of a disease, a disorder or a condition.
The pharmaceutical composition comprising the compound disclosed herein can be administrated via oral, inhalation, rectal, parenteral or topical administration to a subject in need thereof. For oral administration, the pharmaceutical composition may be a regular solid formulation such as tablets, powder, granule, capsules and the like, a liquid formulation such as water or oil suspension or other liquid formulation such as syrup, solution, suspension or the like; for parenteral administration, the pharmaceutical composition may be solution, water solution, oil suspension concentrate, lyophilized powder or the like. Preferably, the formulation of the pharmaceutical composition is selected from tablet, coated tablet, capsule, suppository, nasal spray or injection, more preferably tablet or capsule. The pharmaceutical composition can be a single unit administration with an accurate dosage. In addition, the pharmaceutical composition may further comprise additional active ingredients.
All formulations of the pharmaceutical composition disclosed herein can be produced by the conventional methods in the pharmaceutical field. For example, the active ingredient can be mixed with one or more excipients, then to make the desired formulation. The “pharmaceutically acceptable excipient” refers to conventional pharmaceutical carriers suitable for the desired pharmaceutical formulation, for example: a diluent, a vehicle such as water, various organic solvents, etc, a filler such as starch, sucrose, etc a binder such as cellulose derivatives, alginates, gelatin and polyvinylpyrrolidone (PVP); a wetting agent such as glycerol; a disintegrating agent such as agar, calcium carbonate and sodium bicarbonate; an absorption enhancer such as quaternary ammonium compound; a surfactant such as hexadecanol; an absorption carrier such as Kaolin and soap clay; a lubricant such as talc, calcium stearate, magnesium stearate, polyethylene glycol, etc. In addition, the pharmaceutical composition further comprises other pharmaceutically acceptable excipients such as a decentralized agent, a stabilizer, a thickener, a complexing agent, a buffering agent, a permeation enhancer, a polymer, aromatics, a sweetener, and a dye.
The term “disease” refers to any disease, discomfort, illness, symptoms or indications, and can be interchangeable with the term “disorder” or “condition”.
Abbreviations:
    • AcOH Acetic acid
    • Con. Concentrated
    • D-DBTA (2S, 3S)-Dibenzoyl tartaric acid
    • DCM Dichloromethane
    • DIEA N,N-diisopropylethylamine
    • DMF N,N-dimethylformamide
    • DMF-DMA N,N-dimethylformamide dimethyl acetal
    • DSC Differential Scanning Calorimetry
    • DVS Dynamic Vapor Sorption
    • EtOAc Ethyl Acetate
    • EDCI 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide
    • GC Gas Chromatograph
    • GCMS Gas Chromatography-Mass Spectrometry
    • HATU 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide, hexafluorophosphate
    • HOAc Acetic Acid
    • HOBt Hydroxybenzotriazole
    • HPLC High Performance Liquid Chromatography
    • L-DBTA (2R, 3R)-Dibenzoyl tartaric acid
    • ACN Acetonitrile
    • MeOH Methanol
    • MeMgBr Methyl Magnesium Bromide
    • MsOH Methanesulfonic Acid
    • MTBE Methyl tertiary butyl ether
    • NLT not less than
    • NMP 1-Methyl-2-pyrrolidone
    • NMR Nuclear Magnetic Resonance
    • NMT not more than
    • Pd Palladium
    • RH Relative Humidity
    • RT or rt Room Temperature (20±2° C.)
    • TEA Triethylamine
    • TGA Thermo-gravimetric Analysis
    • THF Tetrahydrofuran
    • XRPD X-ray Powder Diffraction
EXAMPLES
The examples below are intended to be purely exemplary and should not be considered to be limiting in any way. Efforts have been made to ensure accuracy with respect to numbers used (for example, amounts, temperature, etc.), but some experimental errors and deviations should be accounted for. Unless indicated otherwise, temperature is in degrees Centigrade.
The proton nuclear magnetic resonance (1H-NMR) or carbon nuclear magnetic resonance (13C-NMR) spectra was collected on a Agilent instrument operating at 400 MHz. 1HNMR spectra were obtained using CDCl3, CD2Cl2, CD3OD, D2O, d6-DMSO, d6-acetone or (CD3)2CO as solvent and tetramethylsilane (0.00 ppm) or residual solvent (CDCl3: 7.25 ppm; CD3OD: 3.31 ppm; D2O: 4.79 ppm; d6-DMSO: 2.50 ppm; d6-acetone: 2.05; (CD3)2CO: 2.05) as the reference standard. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), q (quartet), qn (quintuplet), sx (sextuplet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, are reported in Hertz (Hz).
The crystalline form is characterized by X-ray powder diffraction (XRPD) pattern method, which is conducted on a PANalytical Empyrean X-ray powder diffractometer with the XRPD parameters as follows:
Parameters for Reflection Mode
X-Ray wavelength Cu, kα, Kα1 (Å): 1.540598, Kα2 (Å): 1.544426
Kα2/Kα1 intensity ratio: 0.50
X-Ray tube setting 45 kV, 40 mA
Divergence slit Automatic
Scan mode Continuous
Scan range (° 2TH) 3°-40°
Step size (° 2TH) 0.0130
Scan speed (°/min) About 10
The thermo-gravimetric analysis (TGA) curves are generated on a TA Q500/Q5000 TGA from TA Instruments. DSC and mDSC curves are generated on a TA Q2000 DSC from TA Instruments. Detailed parameters are listed below:
Parameters TGA DSC mDSC
Temperature range RT-350° C. RT-320° C. 35-150 ° C.
Ramp rate
10° C./min 10° C./min 3° C./min
Purge gas N2 N2 N2
Pan Form Aluminum, Aluminum, Aluminum,
open crimped crimped
The dynamic vapor sorption (DVS) plots are collected via a SMS (Surface Measurement Systems) DVS Intrinsic. The relative humidity at 25° C. is calibrated against deliquescence point of LiCl, Mg(NO3)2 and KCl. Detailed parameters are listed below:
Parameters DVS
Temperature
25° C.
Sample size
10~20 mg
Gas and flow rate N2, 200 mL/min
dm/dt 0.002%/min
Min dm/dt stability duration 10 min
Max. equilibrium time 180 min
RH range
0% RH to 95% RH
RH step size 10% RH from 0% RHto 90% RH
5% RH from 90% RH to 95% RH
Example 1: Preparation of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxy phenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide (Compound 1) Step 1: Synthesis of BG-2
Figure US11555038-20230117-C00005
Under nitrogen atmosphere, TEA (2.4 eq.) was added at 10° C. to a solution of EtOAc (5 v), HOBT (1.2 eq.), EDCI (1.2 eq.), 4-phenoxybenzoic acid (BG-1, 80 Kg, 1.0 eq.) and malononitrile (1.2 eq.). The mixture was then stirred at RT until the reaction was completed. The mixture was then centrifuged and the cake was washed with EtOAc. The filtrate was washed with aqueous NaHCO3 twice and NH4Cl. The organic phase was washed with 1.5 N H2SO4 twice and stirred. The mixture was concentrated, and precipitated from methanol and purified water. The solid was collected by centrifugation and then dried under vacuum to give 79.9 Kg of BG-2. 1H NMR (DMSO-d6) δ 7.62 (d, J=8.6 Hz, 2H), 7.46-7.38 (m, 2H), 7.18 (t, J=7.4 Hz, 1H), 7.06 (d, J=8.0 Hz, 2H), 6.94 (d, J=8.6 Hz, 2H).
Step 2: Synthesis of BG-3
Figure US11555038-20230117-C00006
Under nitrogen atmosphere, a solution of BG-2 (79.9 kg, 1.0 eq.) in ACN (5.0 v) was added into trimethoxymethane (12.0 v) at 85° C. The resultant mixture was stirred until the reaction was completed. The mixture was sampled for HPLC analysis, and concentrated under vacuum. The residue was precipitated from i-PrOH and hexane. The mixture was centrifuged, and the cake was washed with hexane and dried under vacuum. This gave 71.7 Kg of BG-3. 1H NMR (400 MHz, DMSO-d6) δ 7.70 (d, J=8.4 Hz, 2H), 7.52-7.45 (m, 2H), 7.28 (t, J=7.6 Hz, 1H), 7.22-7.06 (m, 4H), 3.93 (s, 3H).
Step 3: Synthesis of BG-4
Figure US11555038-20230117-C00007
Under nitrogen atmosphere, hydrazinium hydroxide (1.0 eq.) in ethanol (0.6 v) was charged dropwise to a solution of BG-3 (71.6 kg, 1.0 eq.) in ethanol (2.5 v) in the reactor below 15° C. The solution was heated to RT and stirred until the reaction was completed. Water (4.0 v) was added to the reactor. The solution was then cooled to 5° C., centrifuged and the cake was washed with water (1.0 v). The cake was dried under vacuum. This gave 66.9 Kg of BG-4. 1H NMR (DMSO-d6) δ 12.11 (br s, 1H), 7.80 (d, J=8.8 Hz, 2H), 7.46-7.39 (m, 2H), 7.18 (t, J=7.6 Hz, 1H), 7.12-7.04 (m, 4H), 6.43 (br s, 2H).
Steps 4 to 6: Synthesis of BG-8
Figure US11555038-20230117-C00008
To a mixture of DCM (8.0 v), BG-5 (80.0 Kg, 1.0 eq.), N,O-dimethylhydroxylamine hydrochloride (1.2 eq.), HOBt (1.2 eq.) and EDCI (1.2 eq.), TEA (2.6 eq.) was charged dropwise below 15° C. the mixture was stirred at RT until the reaction was completed, centrifuged and the cake was washed with DCM (1.0 v) twice. The filtrate was washed with 20% aqueous NH4Cl (3*4.0 v). The filtrate was concentrated under vacuum to give the crude product BG-6, which was used in the next step without further purification. The residue was dissolved in toluene (5.0 v) and THF (1.0 v), cooled to 10° C., charged dropwise MeMgBr (1.4 eq.) at 10° C. and then stirred at RT until the reaction was completed. The solution was cooled below 10° C. Saturated aqueous NH4Cl was charged dropwise below 10° C. The mixture was centrifuged, separated, filtrated, and the organic phase was washed with aqueous NaCl twice. The organic phase was concentrated to give the crude product, which was used in the next step without further purification. The residue in DMF (2.5 v) and DMF-DMA (2.5 v) was stirred at 110° C. until the reaction was completed. The reaction mixture was cooled, concentrated and then DCM was added. The final mixture was washed with saturated aqueous NH4Cl. The organic layer was concentrated and precipitated by charging hexane. The mixture was centrifuged and the cake was collected. The cake was dried under vacuum. This gave 82.2 Kg of the desired product. 1H NMR (DMSO-d6) δ 7.49 (d, J=12.6 Hz, 1H), 5.01 (d, J=12.6 Hz, 1H), 3.99-3.82 (m, 2H), 3.14-2.94 (m, 2H), 2.89-2.61 (m, 6H), 2.49-2.37 (m, 1H), 1.66-1.56 (m, 2H), 1.39 (s, 9H), 1.39-1.20 (m, 2H).
Step 7: Synthesis of BG-9
Figure US11555038-20230117-C00009
Under nitrogen atmosphere, a mixture of toluene (8.0 v), AcOH (0.5 v), BG-8 (1.2 eq.) and BG-4 (66.9 Kg 1.0 eq.) was heated to 95° C. and stirred until the reaction was completed. The mixture was cooled, concentrated and precipitated from methanol. The mixture was centrifuged and the cake was washed with methanol. The cake was dried under vacuum. This gave 107.8 Kg of BG-9. 1H NMR (DMSO-d6) δ 8.78 (d, J=4.6 Hz, 1H), 8.15-8.07 (m, 2H), 7.51-7.41 (m, 2H), 7.34 (d, J=4.6 Hz, 1H), 7.27-7.19 (m, 3H), 7.17-7.10 (m, 2H), 4.24-4.02 (m, 2H), 3.81-3.69 (m, 1H), 3.12-3.82 (m, 2H), 2.15-2.04 (m, 2H), 1.76-1.60 (m, 2H), 1.43 (s, 9H).
Step 8: Synthesis of BG-10
Figure US11555038-20230117-C00010
To a mixture of THF (10.0 v), BG-9 (13.0 Kg, 1.0 eq.) and D-DBTA (1.0 eq.) under N2 was charged Pd/C (10% w/w), hydrogen gas was introduced into the reactor and the hydrogen pressure was maintained to 1.8 MPa. The reactor was heated to 40° C. slowly and stirred until the reaction was completed. The mixture was then cooled, filtered, and the cake was washed with THF. The filtrate was collected, and concentrated under vacuum. DCM was added. The residue was washed with aq. NaHCO3, concentrated and precipitated from MTBE and hexane, then centrifuged. The cake was collected and dried under vacuum to give the desired compound (yield: 94.8% and purity: 98.5%). 1H-NMR (DMSO-d6) δ 7.82-7.76 (m, 2H), 7.56-7.51 (m, 1H), 7.45-7.37 (m, 2H), 7.21-7.14 (m, 1H), 7.12-7.03 (m, 4H), 4.09-3.91 (m, 3H), 3.30-3.22 (m, 2H), 2.82-2.55 (m, 2H), 2.18-1.99 (m, 2H), 1.98-1.86 (m, 1H), 1.69-1.58 (m, 1H), 1.56-1.45 (m, 1H), 1.38 (s, 9H), 1.32-1.13 (m, 2H).
Step 9: Synthesis of BG-11
Figure US11555038-20230117-C00011
To a solution of BG-10 (100.0 Kg 1.0 eq.) in DCM (6.0 v) was added dropwise HCl in EtOH (20.9% w/w, 2.0 v) under nitrogen atmosphere. The mixture is stirred until the reaction was completed. MTBE (4.0 v) was added to the solution, cooled. The cakes was collected by centrifugation and washed with hexane (2.0 V), then the cake was slurried in hexane (5 v), and centrifuged again. The cake was washed with hexane (2.0 V) and dried under vacuum. This gave 85.2 Kg of BG-11. 1H-NMR (DMSO-d6) δ 9.25-8.85 (m, 2H), 7.84-7.70 (m, 2H), 7.47-7.37 (m, 2H), 7.18 (t, J=7.4 Hz, 1H), 7.12-7.03 (m, 4H), 5.73 (br s, 2H), 4.12-4.03 (m, 1H), 3.25-3.19 (m, 4H), 2.90-2.73 (m, 2H), 2.28-2.12 (m, 1H), 2.10-2.00 (m, 1H), 1.99-1.86 (m, 1H), 1.84-1.52 (m, 4H).
Step 10: Synthesis of BG-11A
Figure US11555038-20230117-C00012
A mixture of BG-11 (85.0 Kg, 1.0 eq.) in water (6.0 v) and NaOH (3.0 eq.) was stirred until the reaction was completed at RT. The cake was collected and slurried in MTBE (6.0 v). The mixture was then centrifuged to collect the cake. The cake was dried under vacuum. This gave 71.3 Kg of BG-11A. 1H-NMR (DMSO-d6) δ 7.82-7.74 (m, 2H), 7.54-7.49 (m, 1H), 7.45-7.38 (m, 2H), 7.21-7.14 (m, 1H), 7.12-7.04 (m, 4H), 4.03-3.95 (m, 1H), 3.29-3.21 (m, 2H), 3.00-2.87 (m, 2H), 2.46-2.31 (m, 2H), 2.11-1.83 (m, 3H), 1.58-1.12 (m, 4H).
Step 11: Synthesis of BG-11B
Figure US11555038-20230117-C00013
A mixture of ethanol/water/acetic acid (7:3:1, 46 v) and BG-11A (30 kg, 1.0 eq.) in a reactor was heated to 70±5° C. under nitrogen atmosphere, then a solution of D-DBTA (1.20 eq.) in ethanol/water/acetic acid (7:3:1, 4 v) was added dropwise with the temperature not less than 65° C. The resulting solution was stirred for 16 hrs at 60-65° C., then cooled to RT. The solid was collected by centrifugation and washed with ethanol (2.0 v). The cake was slurried in the mixed solvent of ethanol/water/AcOH (7:3:1, 20 v) for 16 hrs at 55° C. and cooled to RT. The solid was collected by centrifugation, washed with ethanol (2.0 v). The cake was dried under vacuum (Yield: 37.9%) to give the desired product. 1H-NMR (DMSO-d6) δ 8.76 (br s, 2H), 7.99-7.89 (m, 4H), 7.83-7.75 (m, 2H), 7.66-7.57 (m, 3H), 7.52-7.45 (m, 4H), 7.45-7.39 (m, 2H), 7.21-7.14 (m, 1H), 7.13-7.03 (m, 4H), 5.64 (s, 2H), 4.08-4.00 (m, 1H), 3.29-3.19 (m, 4H), 2.85-2.72 (m, 2H), 2.21-1.40 (m, 7H).
Step 12: Synthesis of BG-11C
Figure US11555038-20230117-C00014
To a mixture of dichloromethane (15.0 v) and 20.0% aqueous KOH (3.0 v) was added batchwise BG-11B (48.0 kg, 1.0 eq.) under nitrogen atmosphere at RT. After the reaction was completed, the organic layer was collected and the water layer was extracted with dichloromethane (5.0 v). The organic layers were combined. Con. HCl (0.36 v) was added to the above organic layers at RT. The resulting mixture was stirred until the reaction was completed. The solid was collected by centrifugation and washed with dichloromethane (1.0 v). The collected solid was slurried with MTBE (6.0 v). The solid was collected by centrifugation and washed with MTBE (1.0 v), then was dried under vacuum. This gave 31.5 Kg product (Yield: 100%).
Step 12: Synthesis of BG-11D (Alternative Intermediate)
Figure US11555038-20230117-C00015
ACN (5.0 v), soft water (10.0 v), KOH (5.0 eq.) was charged to a reactor and stirred for at least 15 min. BG-11B (1.0 eq.) was charge to the reactor in portion-wise. The mixture was stirred until the reaction was completed. The cake was collected by centrifugation, slurried in ACN (1.0 v) and soft water (5.0 v), and dried under vacuum to give the product.
Step 13: Synthesis of BG-12
Figure US11555038-20230117-C00016
A solution of BG-11C (15.0 Kg 1.0 eq.) in MsOH (2.5 v) was stirred at 85° C. under nitrogen atmosphere until the reaction was completed. After cooling to 5° C. purified water (4.0 v) was added dropwise to the system and kept the temperature not more than 35° C. (temperature increased obviously). The resulting solution was stirred for 16 hrs at 30° C., and then washed with DCM (2*3.0 v). The aqueous phase was collected. DCM (6.0 v) was added to the aqueous phase, the mixture was cooled to 5° C. The pH value was adjusted to 11-12 with 20% aqueous NaOH (temperature increased obviously) with stirring with the temperature not more than 30° C. The organic phase was separated and collected. The aqueous phase was extracted with DCM (3.0 v). The organic layers were combined and concentrated. MTBE (4.0 v) was added to the residue. The mixture was then concentrated and precipitated from n-heptane. The solid was collected by centrifugation and dried in a vacuum oven. This gave 12.55 Kg of BG-12 (Yield: 94.9%). 1H-NMR (DMSO-d6) δ 7.52-7.46 (m, 2H), 7.45-7.38 (m, 2H), 7.21-7.13 (m, 1H), 7.12-7.03 (m, 4H), 6.64 (s, 1H), 3.99-3.90 (m, 1H), 3.29-3.22 (m, 2H), 3.03-2.90 (m, 2H), 2.48-2.36 (m, 2H), 2.03 (dd, J=13.9, 5.6 Hz, 2H), 2.14-1.99 (m, 1H), 1.97-1.85 (m, 1H), 1.65-1.15 (m, 3H).
Step 14: Synthesis of BG-13
Figure US11555038-20230117-C00017
A mixture of MeOH (13.5 v), purified water (4.5 v) and BG-12 (8.5 Kg, 1.0 eq.) in a reactor was heated to 50° C. under N2 atmosphere. To the mixture was charged dropwise a solution of L-DBTA (0.7 eq.) in MeOH/purified water (1.5 v/0.5 v) while keeping the temperature at 50° C. After addition, the mixture was stirred for at least 2 hrs at 50° C., and then cooled to RT and stirred for at least 16 hrs at RT. The cake was collected by centrifugation and was washed with MeOH (2.0 v). The cake was dried in a vacuum oven. This gave 9.08 Kg of BG-13 (Yield: 74.8%).
Step 15: Synthesis of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide (Compound 1)
Figure US11555038-20230117-C00018
Under N2 atmosphere, BG-13 (6.0 Kg; 1.0 eq) was stirred in charged DCM (30 L; 5.0 v) with the presence of 5% w/w aq. NaOH (0.48 Kg; 1.2 eq.) at around 30° C. until the reaction was completed. Separated and collected organic layer. The aqueous layer was further extracted with DCM. All organic layers were combined and washed with 15% brine for twice.
The organic layer was collected and concentrated under vacuum. To the residue was added but-2-ynoic acid (0.89 Kg; 1.05 eq.), HATU (4.21 Kg; 1.1 eq.) under N2 atmosphere. The mixture was heated to 35˜40° C. To the mixture was added dropwise TEA (3.05 Kg; 3.0 eq.; dissolved with DCM (12.0 L; 2.0 v)). The reaction was stirred until the reaction was completed.
The reaction system was concentrated under vacuum and swapped to DMF solution. Water (506.8 L; 40.0 v) was added dropwise to precipitate the solid. Centrifuged, collected the cake. The cake was dissolved in DCM and washed with 8% NaHCO3 and 15% brine to remove DMF residue. The organic layer was concentrated and purified by silica gel (100-200 mesh column, eluted with heptane, followed by 1.2% w/w % methanol in DCM). The solution of Compound 1 was collected, concentrated and precipitate from DMF and water. The residual was centrifuged, and collected to give the crude product of Compound 1, which was confirmed to be amorphous.
Example 2: Preparation of Form A of Compound 1
The crude product of Compound 1 prepared in the above Example 1 was dissolved in 5.0 volumes of DCM. The resultant solution was washed with water (3.0 volumes*2). The organic phase was concentrated and swapped to EtOAc (no more than 2 volumes). The solution was further charged with EtOAc to 4.5 volumes. 27.5% w/w n-heptane in EtOAc (12.0 v) (the volume/volume ratio of EtOAc:n-heptane is about 2:1) was added dropwise to the system while the temperature was kept at RT. The system was then stirred for at least 24 hours at RT, centrifuged and collected the cake to obtain the resultant product in solid form.
The solid was then subject to various characterizations including XRPD (FIG. 2A), TGA/DSC (FIG. 2B) and 1H NMR (FIG. 2C). 1H-NMR (DMSO-d6) δ 7.56-7.47 (m, 2H), 7.46-7.37 (m, 2H), 7.22-7.13 (m, 1H), 7.13-7.02 (m, 4H), 6.67 (s, 1H), 4.45-4.20 (m, 2H), 4.09-3.95 (m, 1H), 3.33-3.26 (m, 2H), 3.17-2.95 (m, 1H), 2.70-2.52 (m, 1H), 2.36-2.18 (m, 1H), 2.00 (s, 3H), 2.12-1.85 (m, 2H), 1.83-1.66 (m, 1H), 1.65-1.51 (m, 1H), 1.37-1.06 (m, 2H).
The XRPD results found the resultant solid as a crystalline form (designated as Form A). The DSC result showed an endotherm at 87.5° C. before decomposition at 274.8° C. The TGA result showed a weight loss of 8.42% up to 150° C. After desolvation via heating in TGA, amorphous was produced (FIG. 2D). The 1H NMR result showed an EtOAc content of around 9.1%, consistent with TGA weight loss. The above 1H-NMR and TGA heating confirmed Form A as an EtOAc solvated crystalline form (sometimes referred to as Form A as an EtOAc solvate) wherein the molar ratio of Compound 1 and EtOAc is about 2:1.
Example 3: Preparation of Other Form A of Compound 1
About 15 mg of Form A as an EtOAc solvate as prepared in Example 2 was dissolved in the respective solvent (0.40 mL EtOH or 0.15 mL acetone) to obtain a saturated solution at room temperature. The resultant solution was magnetically stirred, then followed by addition of n-heptane as the anti-solvent (2.4 mL n-heptane for EtOH and 0.4 mL n-heptane for acetone, respectively) to induce precipitation. The respective precipitate was isolated for XRPD analysis.
The XRPD analysis found the product obtained from crystallization from EtOH/n-heptane and the product obtained from crystallization from acetone/n-heptane are crystalline; see FIG. 3A.
It is interesting to note that FIG. 3A shows that the XRPD patterns of Form A obtained from 2:1 EtOAc/n-heptane (v/v) in Example 2, Form A obtained from EtOH/n-heptane and Form A obtained from acetone/n-heptane comprise the diffraction peaks of substantially the same positions, although their relative intensities vary.
Alternatively, Form A with increased crystallinity was prepared by slurry of amorphous in EtOH/n-heptane (1:3, v/v) at RT, as shown in FIG. 3B (also confirmed the crystalline structure of Form A, referred to as Form A obtained from slurry conversion of EtOH/n-heptane). The TGA curve of Form A obtained from slurry conversion of EtOH/n-heptane in FIG. 3C showed a two-step loss of 6.8% up to 150° C. The 1H-NMR result of Form A obtained from slurry conversion of EtOH/n-heptane in FIG. 3D showed an EtOH content of 0.4% and n-heptane content of 4.4% without EtOAc residual. After desolvation via TGA heating, amorphous was also produced as shown in FIG. 3E.
The 1H-NMR and TGA heating for Form A obtained from slurry conversion from EtOH/n-heptane also confirmed the resultant Form A as a solvated crystalline form, in particular, Form A as a hetero-solvate of EtOH and n-heptane.
Example 4: Preparation Crystalline Form B of Compound 1
To the reactor was charged Form A of Compound 1 (1.0 eq.) prepared in the above Example 2, EtOAc (7.5 v) and n-heptane (7.5 v). The mixture was heated to 55-60° C. and stirred at least 72 h. The mixture was then cooled to 10-15° C. and stirred for at least 15 min. The cake was collected after centrifugation and washed with n-heptane to give a solid (yield 52.2%).
The resultant solid was then subject to various characterizations including XRPD (FIG. 4A), TGA/DSC (FIG. 4B), 1H NMR (FIG. 4C) and 13C-NMR (DMSO-d6) (FIG. 4D). 1H-NMR (DMSO-d6) δ 7.56-7.47 (m, 2H), 7.46-7.37 (m, 2H), 7.22-7.13 (m, 1H), 7.13-7.02 (m, 4H), 6.67 (s, 1H), 4.45-4.20 (m, 2H), 4.09-3.95 (m, 1H), 3.33-3.26 (m, 2H), 3.17-2.95 (m, 1H), 2.70-2.52 (m, 1H), 2.36-2.18 (m, 1H), 2.00 (s, 3H), 2.12-1.85 (m, 2H), 1.83-1.66 (m, 1H), 1.65-1.51 (m, 1H), 1.37-1.06 (m, 2H).
The XRPD result found the resultant solid as a crystalline form different from the above Form A (designated as Form B). The DSC result showed a neat melting endotherm at 153.9° C. (onset temperature). The TGA result showed a two-step weight loss of 2.3% up to 140° C., indicating Form B is anhydrous non-solvated. The 1H-NMR showed no solvent residual EtOAc that appeared in Form A, which is consistent with the TGA result.
Example 5: Preparation of Crystalline Form B of Compound 1 Example 5A: Preparation from Form A of Compound 1
250 mg of Form A of Compound 1 prepared in the above Example 2 was charged into a 20-mL glass vial. 2.0 mL of THF was added into the vial and stirred at RT until all solids were dissolved. 2.0 mL of n-heptane was added to induce precipitate and the mixture was equilibrated at RT for 1 hr. About 4 mg of Form B prepared in the above Example 4 was added as seed and the mixture was stirred at RT for 1.5 hrs. Additional 4.0 mL of n-heptane was added to induce more solids and transfer to 50° C. agitation for 1 hr. About 4 mg of Form B prepared in the above Example 4 was added again as seed and the mixture was stirred at 50° C. for 22 hrs. The cake was then collected by vacuum filter and was dried at 50° C. for 16 hrs to obtain the solid (169.5 mg, yield of ˜74%).
Example 5B: Preparation from Compound 1 in Amorphous Form
250 mg of Compound 1 in amorphous form prepared in Example 1 was added into a 20-mL glass vial. 1.0 mL of THF was added into the vial and stirred at RT until all solids were dissolved. 1.0 mL of n-heptane was added to induce precipitate. The mixture was equilibrated at RT for 1 hr. About 4 mg of Form B of Compound 1 prepared in the above Example 4 was added as seed and the mixture was stirred at RT for 1.5 hrs. Additional 2.0 mL of n-heptane was added to induce more solids. About 4 mg of Form B of Compound 1 prepared in the above Example 4 was added again as seed and the mixture was stirred at 50° C. for 22 hrs. The cake was collected by vacuum filter and dried at 50° C. for 16 hrs to obtain the solid (168.2 mg, yield of ˜72%).
The XRPD analysis of the resultant solids of Examples 5A and 5B confirmed that the solids of Example 5A and 5B are Form B as their XRPD patterns (FIG. 4G) are consistent with that of Example 4 (FIG. 4A).
Example 6: Slurry Conversion from Compound 1 of Form A or Amorphous Form at Different Temperatures
Slurry conversion experiments were conducted in different solvent systems at different temperature with Compound 1 in Form A or Compound 1 in amorphous form as the starting material.
Example 6A: Slurry Conversion from Form A at 5° C.
Slurry conversion experiments were conducted in different solvent systems at 5° C. by suspending about 15 mg of Form A prepared in the above Example 2 with 0.3 mL of solvent in a 1.5-mL glass vial. After stirring for about 6 days, the suspensions were isolated by centrifuge, and the solids were analyzed by XRPD. Results summarized in Table 3 showed Form A was obtained from some solvent systems at 5° C., as the XRPD patterns of the resultant crystalline forms are consistent with Form A as an EtOAc solvate in Example 2.
TABLE 3
Summary of slurry conversion experiments at 5° C.
No. Solvent (v/v) Solid Form
A01 Isopropyl acetate Form A
A02 Isopropyl alcohol N/A
A03 H2O Form A
A04 MTBE Form A
A05 n-heptane Form A
A06 MeOH/H2O (l:5) amorphous
A07 ACN/H2O (1:5) N/A
A08 Acetone/H2O (1:5) amorphous
A09 Acetic acid/H2O (1:5) N/A
A10 NMP/H2O (1:5) Form A
A11 THF/n-heptane(l:5) Form A
A12
1,4-Dioxane/n-heptane(l:5) Form A
A13 EtOAc/n-heptane(l:5) Form A
A14 DCM/MTBE(1:5) Form A
N/A: no solid was obtained.
Example 6B: Slurry Conversion from Form A at RT
Slurry conversion experiments were conducted in different solvent systems at RT by suspending about 15 mg of Form A with 0.5 mL of solvent in a 1.5-mL glass vial. After stirring for about 6 days, the suspensions were isolated by centrifuge, and the solids were analyzed by XRPD. Results summarized in Table 4 showed Form A was obtained from some solvent systems at RT, as the XRPD patterns of the resultant crystalline forms are consistent with Form A as an EtOAc solvate in Example 2.
TABLE 4
Summary of slurry conversion experiments at RT
No. Solvent (v/v) Solid Form
B0l Isopropyl acetate Form A
B02 Isopropyl alcohol N/A
B03 H2O amorphous
B04 MTBE Form A
B05 MeOH/H2O(l:5) amorphous
B06 ACN/H2O (1:5) N/A
B07 Acetone/H2O (1:5) N/A
B08 Acetic acid/H2O (1:5) N/A
B09 NMP/H2O (1:5) amorphous
B10 THF/n-heptane(l:5) Form A
B11
1,4-dioxane/n-heptane(l:5) Form A
B12 EtOAc/n-heptane(l:5) Form A
B13 CHCl3/MTBE(1:5) Form A
N/A: no solid was obtained.
Example 6C: Slurry Conversion from Compound 1 in Amorphous Form
Slurry conversion experiments were conducted in varying solvent systems at RT and 50° C. using Compound 1 in amorphous form as the staring material. A certain amount (10-25 mg) of Compound 1 in amorphous form was added into the corresponding solvent. The resultant mixture was stirred at the desired temperature for 2 hrs. About 2 mg of Form B was added as seed. The mixture was stirred for another 60 hrs at the same desired temperature. The resultant solid was collected for XRPD analysis.
TABLE 5
Summary of slurry conversion experiments starting
from Compound 1 in amorphous form
No. Solvent (v/v) Condition Solid Form
C01 EtOAc/n-hexane (1:1) RT, Form A + B
C02 EtOAc/n-hexane (1:3) Form B seeded Form B
C03 EtOH/n-heptane (1:3) Form A
C04 Acetone/n-heptane (1:3) Form A + B
C05 THF/n-heptane (1:3) Form A + B
C06 CHCl3/n-heptane (1:3) Gel
C07 EtOAc/n-hexane (1:1) 50° C., Form B
C08 EtOAc/n-hexane (1:3) Form B seeded Form B
C09 EtOH/n-heptane (1:3) Form A
C10 Acetone/n-heptane (1:3) Form B
C11 THF/n-heptane (1:3) Form B
C12 CHCl3/n-heptane (1:3) Form B
C13 EtOH/H2O (1:5) Gel
C14 THF/H2O (1:5) Gel
C15 CHCl3/MTBE (1:5) Form B
C16 Acetone/MTBE (1:5) Form B
The above table showed that Compound 1 in amorphous form can be converted into Form A, Form B or mixture thereof in slurry conversion conducted in varying solvent systems at room temperature (20±2° C.) and 50° C. followed by addition of Form B seeds. FIG. 5A and FIG. 5B confirmed the crystalline forms of the resultant solids obtained at different temperatures.
Example 6D: Thermodynamic Stability in Current Process Solvent Systems
About 500 mg, 300 mg, 100 mg and 70 mg of Compound 1 in amorphous form of Example 1 were weighted into each 3-mL glass vial. 1.0 mL of EtOAc/n-heptane (solvent ratio: 1/0*, 3/1, 1/1, 1/3) was added into each of the above vial sequentially. The mixtures were stirred at 50° C. for 2 hrs to get saturated solutions. Equally, about 10 mg of Compound 1 in Form A of Example 2 and Form B of Example 4 in an approximate mass ratio of 1:1 were added into each 1.5 mL glass vial. 0.3 mL of each of the above corresponding saturated solution (equilibrated at 50° C.) was added into each of the above 1.5 mL glass vial and stirred at desired temperature. Samples were collected after 3, 6 and 13 days for XRPD characterization until form conversion was observed. (*Clear solution in pure EtOAc was obtained due to not enough Compound 1 in amorphous form)
Results were summarized in Table 6, FIGS. 5C, 5D and 5E.
TABLE 6
Summary of thermodynamic stability
in current process solvent systems
No. Solvent (v/v) Condition Solid Form
D01 EtOAc
10° C. Form A
D02 EtOAc/n-heptane (3:1) Form A
D03 EtOAc/n-heptane (1:1) Form A
D04 EtOAc/n-heptane (1:3) Form A + B
D05 EtOAc RT Form A
D06 EtOAc/n-heptane (3:1) Form A
D07 EtOAc/n-heptane (1:1) Form A
D08 EtOAc/n-heptane (1:3) Form A* + B
D09 EtOAc
50° C. Form B
D10 EtOAc/n-heptane (3:1) Form B
D11 EtOAc/n-heptane (1:1) Form B
D12 EtOAc/n-heptane (1:3) Form B
*Small part of Form A was observed, indicating Form B is thermodynamically more stable under this condition.
In the above competitive slurry turnover experiments, Form A and Form B were exposed under EtOAc/n-heptane system with a series of solvent ratios and temperatures (10° C., 20±2° C., and 50° C.), which indicated that Form B is thermodynamically more stable than Form A at 50° C., and at room temperature with a volume fraction of EtOAc decreased to 0.25. Concluded from Example 6, anhydrous non-solvated Form B tends to be the stable crystalline form under conditions with elevated temperature, decreased EtOAc content, seed loading and extended equilibration time.
Example 7: Hygroscopic Test
Compound 1 in Form B prepared in Example 4 was subjected to hygroscopic test using dynamic vapor sorption (DVS). The DVS plots is shown in FIG. 4E suggesting Form B is non-hygroscopic. After the DVS test, no form change was observed by XRPD comparison as shown in FIG. 4F.
The long term stability studies of Compound 1 in Form B showed there was no significant chemical purity change occurred when stored at 25° C./60% RH for up to 12 months and at 40° C./75% RH condition for up to 6 months. In addition, no crystal form and optical purity changes were observed when stored at 25° C./60% RH for up to 12 months and at 40° C./75% RH condition for up to 6 months.
Example 8: Mechanical Test
Grinding experiments were conducted in two conditions with water and without any solvent. About 15 mg of Compound 1 in Form A prepared in Example 2 was added into the mortar and manually ground with/without solvents for 5 minutes before the solids were collected and analyzed. As summarized in Table 7, amorphous was obtained by XRPD characterization.
TABLE 7
Summary of grinding experiments
No. Solvent Solid Form
1 N/A amorphous
2 H2O amorphous
N/A: no solvent was added.
Grinding experiments were also conducted with respect to Form B of Example 4 to evaluate possible form change of Form B under mechanical force. Neat and water-assisted grinding experiments were performed with respect to Form B. About 15 mg of Compound 1 in Form B prepared in Example 4 was added into the mortar and manually ground with/without water for 5 minutes. Then solids were collected for XRPD characterization. FIG. 4H showed that mechanical strain did not change Form B to become amorphous or other forms, although crystallinity has been decreased after grinding. The result further confirmed Form B is more stable then Form A.
Example 9
Determination of Absolute Configuration of Compound 1
Preparation of BG-13 Single Crystal
Six single crystal growth experiments (see Table 8) were performed via slow cooling. Suitable single crystals of BG-13 were obtained by slow cooling in MeOH/H2O (1:1, v/v).
TABLE 8
Single Crystal Growth Experiments
Experiment Weight Solvent Temperature Dissolved
ID (mg) (1 mL, v/v) Method (° C.) (Y/N*) Observation
1 5.6 IPA/H2O (3/1) cooling 60 N Block-like crystal
2 5.5 IPA/H2O (3/1) cooling 60 N Block -like crystal
3 5.4 IPA/H2O (3/1) cooling 60 N Block -like crystal
4 5.5 IPA/H2O (3/1) cooling 60 N Block -like crystal
5 4.7 MeOH/H2O (2/1) cooling 60 N Crystal
6 5.5 MeOH/H2O (1/1) cooling 60 N Crystal
The single crystal data and structure refinement data were generated as shown in Table 9 on a Bruker APEX DUO single-crystal diffractometer with CCD detector (Cu Kα, λ=1.54178 Å, 173.15 K).
TABLE 9
Single Crystal Data and Structure Refinement of BG-13
Empirical formula C33H34N5O6
Formula weight 596.65
Temperature 173.15
Wavelength 1.54178 Å
Crystal system, space monoclinic C2
group
Unit cell dimensions a = 16.7939(4) Å alpha = 99.00 deg..
b = 7.9871(2) Å beta = 108.0460(10) deg.
c = 23.5438(5) Å gamma = 90.00 deg.
Volume 3002.69(12) Å3
Z, Calculated density 4 1.320 mg/mm3
Absorption coefficient 0.756 mm−1
F(000) 1260.0
Crystal size 0.3 × 0.21 × 0.08 mm3
Theta range for data 1.97 to 64.96 deg.
collection
Limiting indices −19 <= h <= 1,
−7 <= k <= 9,
−27 <= l <= 24
Reflections 5073/3756
collected/unique [R(int) = 0.1062]
Completeness 92.8%
Refinement method Full matrix least
squares on F2
Data/restraints/parameters 3756/1/398
Goodness-of-fit on F2 1.192
Final R indices R1 = 0.0819 wR2 = 8.2294
[I > 2sigma(I)]
Absolute structure Flack 0.0(3)
Largest diff. peak and 0.50 and −0.57 e · A−3
hole
BG-13 was confirmed to be a (2R, 3R)-dibenzoyl tartaric acid (L-DBTA) salt and the molar ratio of freebase to L-DBTA is 2:1. Configuration of both carbons (C32 and C32′) in L-DBTA was confirmed to be R. Configuration of C6 in freebase was determined to be S, as shown in FIG. 6A to FIG. 6C. A powder X-ray diffraction pattern method was also used to characterize the structure of the single crystals, as shown in FIG. 6D.
Absolute Configuration of Compound 1
The absolute configurations of Compound 1 was deduced to be S from the single crystal X-ray structural analysis of intermediate BG-13.
EFFICACY TESTS
(1) BTK Kinase Enzymatic Assay
Form B of Compound 1 was tested for inhibition of BTK kinase (aa2-659, Carna Biosciences) in assays based on the time-resolved fluorescence-resonance energy transfer (TR-FRET) methodology. The assays were carried out in 384-well low volume black plates in a reaction mixture containing BTK kinase, 5 μM ATP, 2 μM peptide substrate and 0-10 μM Form B of Compound 1 of Example 4 in buffer containing 50 mM Tris pH7.4, 10 mM MgCl2, 2 mM MnCl2, 0.1 mM EDTA, 1 mM DTT, 0.005% Tween-20, 20 nM SEB and 0.01% BSA. The kinase was incubated with compound for 60 minutes at room temperature and the reaction was initiated by the addition of ATP and peptide substrate. After reaction at room temperature for 60 minutes, an equal volume of stop/detection solution was added according to the manufacture's instruction (CisBio Bioassays). The stop/detection solution contained Eu3+ cryptate-conjugated mouse monoclonal antibody (PT66) anti-phosphotyrosine and XL665-conjugated streptavidin in buffer containing 50 mM HEPES pH 7.0, 800 mM KF, 20 mM EDTA, and 0.1% BSA. Plates were sealed and incubated at room temperature for 1 hour, and the TR-FRET signals (ratio of fluorescence emission at 665 nm over emission at 620 nm with excitation at 337 nm wavelength) were recorded on a PHERAstar FS plate reader (BMG Labtech). Phosphorylation of peptide substrate led to the binding of anti-phosphotyrosine antibody to the biotinylated peptide substrate, which places fluorescent donor (Eu3+ crypate) in close proximity to the accepter (Streptavidin-XL665), thus resulting in a high degree of fluorescence resonance energy transfer from the donor fluorophore (at 620 nm) to the acceptor fluorophore (at 665 nm). Inhibition of BTK kinase activity resulted in decrease of the TR-FRET signal. The IC50 for Compound 1 in Form B was derived from fitting the data to the four-parameter logistic equation by Graphpad Prism software.
The IC50 of Compound 1 in Form B is 1 nM.
(2) Biochemical Kinase Selectivity
Selectivity of Form B was profiled against a panel of 367 kinases at 2 μM at Reaction Biology Corp. Form B displayed less than 70% inhibition against 358 kinases, and greater than 70% inhibition against 9 kinases including BTK.

Claims (7)

The invention claimed is:
1. Crystalline Form A of Compound 1:
Figure US11555038-20230117-C00019
wherein the crystalline form is an ethyl acetate solvate; and
wherein the crystalline form is characterized by an X-ray powder diffraction pattern comprising diffraction peaks having angle values (° 2θ) at 18.6°±0.2° 2θ, 19.8°±0.2° 2θ, and 20.6°±0.2° 2θ.
2. The crystalline form of claim 1, wherein the crystalline form is further characterized by an X-ray powder diffraction pattern comprising additional diffraction peaks having angle values (° 2θ) at 5.3°±0.2° 2θ and 16.90±0.2° 2θ.
3. The crystalline form of claim 1, wherein the crystalline form is further characterized by an X-ray powder diffraction pattern comprising additional diffraction peaks having angle values (° 2θ) at 5.3°±0.2° 2θ, 10.9°±0.2° 2θ, 16.9°±0.2° 2θ, 21.1°±0.2° 2θ, 22.5°±0.2° 2θ, and 22.8°±0.2° 2θ.
4. The crystalline form of claim 1, wherein the crystalline form is further characterized by an X-ray powder diffraction pattern comprising additional diffraction peaks having angle values (° 2θ) at 5.3°±0.2° 2θ, 10.9°±0.2° 2θ, 11.2°±0.2° 2θ, 13.4°±0.2° 2θ, 14.4°±0.2° 2θ, 16.9°±0.2° 2θ, 21.1°±0.2° 2θ, 21.7°±0.2° 2θ, 22.5°±0.2° 2θ, 22.8°±0.2° 2θ, 23.6°±0.2° 2θ, and 24.3°±0.2° 2θ.
5. The crystalline form of claim 1, wherein the crystalline form is further characterized by a differential scanning calorimetry curve in accordance with FIG. 2B.
6. The crystalline form of claim 1, wherein the crystalline form is further characterized by a thermogravimetric analysis curve in accordance with FIG. 2B.
7. A process for preparing crystalline Form A of Compound 1 of claim 1:
Figure US11555038-20230117-C00020
wherein the crystalline form is an ethyl acetate solvate;
wherein the crystalline form is characterized by an X-ray powder diffraction pattern comprising diffraction peaks having angle values (° 2θ) at 18.6°±0.2° 2θ, 19.8°±0.2° 2θ, and 20.6°±0.2° 2θ; and
wherein the process comprises the following steps:
(a) dissolving an amorphous form of Compound 1 in a solvent selected from the group consisting of acetic acid, acetone, acetonitrile, chloroform, dichloromethane, N,N-dimethylacetamide, dimethylsulfoxide, 1,4-dioxane, ethanol, ethyl acetate, isopropanol, isopropyl acetate, methanol, methyl isobutyl ketone, 1-methyl-2-pyrrolidone, tetrahydrofuran, 2-methyltetrahydrofuran, and toluene, or a mixture thereof;
(b) adding an anti-solvent selected from the group consisting of cyclohexane, n-heptane, n-hexane, methyl tert-butyl ether, petroleum ether, and water, or a mixture thereof, to the solution provided in step (a), to induce precipitation; and
(c) maintaining the mixture provided in step (b) at a temperature below 25° C. or at a temperature of 25° C., to precipitate the crystalline Form A of Compound 1 of claim 1.
US16/479,709 2017-01-25 2018-01-25 Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof Active 2039-07-13 US11555038B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
WOPCT/CN2017/072553 2017-01-25
CNPCT/CN2017/072553 2017-01-25
CN2017072553 2017-01-25
PCT/CN2018/074108 WO2018137681A1 (en) 2017-01-25 2018-01-25 Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a] pyrimidine-3-carboxamide, preparation, and uses thereof

Publications (2)

Publication Number Publication Date
US20210332049A1 US20210332049A1 (en) 2021-10-28
US11555038B2 true US11555038B2 (en) 2023-01-17

Family

ID=62978113

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/479,709 Active 2039-07-13 US11555038B2 (en) 2017-01-25 2018-01-25 Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof

Country Status (5)

Country Link
US (1) US11555038B2 (en)
EP (1) EP3573989A4 (en)
CN (1) CN110461847B (en)
TW (1) TWI774726B (en)
WO (1) WO2018137681A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11701357B2 (en) 2016-08-19 2023-07-18 Beigene Switzerland Gmbh Treatment of B cell cancers using a combination comprising Btk inhibitors
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
US11851437B2 (en) 2016-08-16 2023-12-26 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6204568B2 (en) 2013-04-25 2017-09-27 ベイジーン,リミテッド Fused heterocyclic compounds as protein kinase inhibitors
MX2016003292A (en) 2013-09-13 2016-06-24 Beigene Ltd Anti-pd1 antibodies and their use as therapeutics and diagnostics.
JP6526189B2 (en) 2014-07-03 2019-06-05 ベイジーン リミテッド Anti-PD-L1 antibodies and their use for therapy and diagnosis
CN109475536B (en) 2016-07-05 2022-05-27 百济神州有限公司 Combination of a PD-l antagonist and a RAF inhibitor for the treatment of cancer
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
AU2018290532A1 (en) 2017-06-26 2019-11-21 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
CN110997677A (en) 2017-08-12 2020-04-10 百济神州有限公司 Btk inhibitors with improved dual selectivity
CA3160368A1 (en) * 2019-12-04 2021-06-10 Henan Zhiwei Biomedicine Co., Ltd. Substituted imidazolecarboxamide as bruton's tyrosine kinase inhibitors
US11786531B1 (en) 2022-06-08 2023-10-17 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder

Citations (198)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1412017A (en) 1971-12-09 1975-10-29 Icn Pharmaceuticals Pyrazolo 1,5a pyrimidines
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
JPH07278148A (en) 1994-11-18 1995-10-24 Sankyo Co Ltd Imidazopyrazole derivative
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5629204A (en) 1994-03-01 1997-05-13 Ono Pharmaceutical Co., Ltd. Peptide related to human programmed cell death and DNA encoding it
US5994514A (en) 1991-08-14 1999-11-30 Genentech, Inc. Immunoglobulin variants
US6165745A (en) 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO2001016138A1 (en) 1999-08-27 2001-03-08 Abbott Laboratories Sulfonylphenylpyrazole compounds useful as cox-2 inhibitors
WO2001019829A2 (en) 1999-09-17 2001-03-22 Basf Aktiengesellschaft Pyrazolopyrimidines as therapeutic agents
WO2002050071A1 (en) 2000-12-21 2002-06-27 Bristol-Myers Squibb Company Thiazolyl inhibitors of tec family tyrosine kinases
US20020094989A1 (en) 2000-10-11 2002-07-18 Hale Jeffrey J. Pyrrolidine modulators of CCR5 chemokine receptor activity
WO2002072576A1 (en) 2001-03-09 2002-09-19 Pfizer Products Inc. Benzimidazole anti-inflammatory compounds
WO2003004497A1 (en) 2001-07-05 2003-01-16 Sumitomo Pharmaceuticals Company, Limited Novel heterocyclic compound
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6632927B2 (en) 1989-12-21 2003-10-14 Celltech Therapeutics Limited Humanized antibodies
US20040038339A1 (en) 2000-03-24 2004-02-26 Peter Kufer Multifunctional polypeptides comprising a binding site to an epitope of the nkg2d receptor complex
WO2004017908A2 (en) 2002-08-26 2004-03-04 Takeda Pharmaceutical Company Limited Calcium receptor modulating compound and use thereof
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6808710B1 (en) 1999-08-23 2004-10-26 Genetics Institute, Inc. Downmodulating an immune response with multivalent antibodies to PD-1
WO2005005429A1 (en) 2003-06-30 2005-01-20 Cellular Genomics, Inc. Certain heterocyclic substituted imidazo[1,2-a]pyrazin-8-ylamines and methods of inhibition of bruton’s tyrosine kinase by such compounds
WO2005011597A2 (en) 2003-07-29 2005-02-10 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2005014599A1 (en) 2003-06-04 2005-02-17 Cellular Genomics, Inc. Imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of bruton’s tyrosine kinase by such compounds
US20050064514A1 (en) 2003-01-09 2005-03-24 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2005047290A2 (en) 2003-11-11 2005-05-26 Cellular Genomics Inc. Imidazo[1,2-a] pyrazin-8-ylamines as kinase inhibitors
WO2005077981A2 (en) 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES
US6936704B1 (en) 1999-08-23 2005-08-30 Dana-Farber Cancer Institute, Inc. Nucleic acids encoding costimulatory molecule B7-4
US20050202028A1 (en) 2001-01-17 2005-09-15 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
CN1753912A (en) 2002-12-23 2006-03-29 惠氏公司 Antibodies against PD-1 and uses therefor
US7029674B2 (en) 2001-04-02 2006-04-18 Wyeth Methods for downmodulating immune cells using an antibody to PD-1
CN1771231A (en) 2002-08-26 2006-05-10 武田药品工业株式会社 Calcium receptor modulating compound and use thereof
WO2006053121A2 (en) 2004-11-10 2006-05-18 Cgi Pharmaceuticals, Inc. Imidazo[1 , 2-a] pyrazin-8-ylamines useful as modulators of kinase activity
US20060121032A1 (en) 2003-03-03 2006-06-08 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
WO2006065946A1 (en) 2004-12-16 2006-06-22 Vertex Pharmaceuticals Incorporated Pyrid-2-ones useful as inhibitors of tec family protein kinases for the treatment of inflammatory, proliferative and immunologically-mediated diseases
US20060134105A1 (en) 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
WO2006084015A2 (en) 2005-02-04 2006-08-10 Genentech, Inc. Raf inhibitor compounds and methods
US20060183746A1 (en) 2003-06-04 2006-08-17 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of Bruton's tyrosine kinase by such compounds
WO2006099075A2 (en) 2005-03-10 2006-09-21 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
US20060263856A1 (en) 2001-03-07 2006-11-23 Emd Lexigen Research Center Corp. Expression technology for proteins containing a hybrid isotype antibody moiety
WO2006133396A2 (en) 2005-06-08 2006-12-14 Dana-Farber Cancer Institute Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007026720A1 (en) 2005-08-31 2007-03-08 Taisho Pharmaceutical Co., Ltd. Ring-fused pyrazole derivative
WO2007026950A1 (en) 2005-09-01 2007-03-08 Astellas Pharma Inc. Pyridazinone derivatives used for the treatment of pain
WO2007027729A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3, 5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
WO2007027594A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
WO2007067444A1 (en) 2005-12-08 2007-06-14 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
US20070160597A1 (en) 2002-03-01 2007-07-12 Xencor, Inc. Optimized Fc variants and methods for their generation
WO2007087068A2 (en) 2006-01-13 2007-08-02 Pharmacyclics, Inc. Inhibitors of tyrosine kinases and uses thereof
WO2007136572A2 (en) 2006-05-15 2007-11-29 Merck & Co., Inc. Antidiabetic bicyclic compounds
WO2007136790A2 (en) 2006-05-18 2007-11-29 Mannkind Corporation Intracellular kinase inhibitors
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
CN101104640A (en) 2006-07-10 2008-01-16 苏州大学 Preparation for anti human PD-L1 monoclonal antibody and application thereof
WO2008033854A1 (en) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
WO2008033857A2 (en) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Substituted amides, method of making, and method of use thereof
WO2008033834A1 (en) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Pyrimidines derivatives and their use as kinase inhibitors
US20080076921A1 (en) 2006-09-22 2008-03-27 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2008054827A2 (en) 2006-11-03 2008-05-08 Pharmacyclics, Inc. Bruton's tyrosine kinase activity probe and method of using
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US7414171B2 (en) 2000-11-15 2008-08-19 Ono Pharmaceutical Co., Ltd. PD-1-lacking mouse and use thereof
US7416726B2 (en) 2000-04-13 2008-08-26 The Rockefeller University Enhancement of antibody-mediated immune responses
WO2008144253A1 (en) 2007-05-14 2008-11-27 Irm Llc Protein kinase inhibitors and methods for using thereof
WO2008145142A1 (en) 2007-05-31 2008-12-04 Genmab A/S Stable igg4 antibodies
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2009039397A2 (en) 2007-09-20 2009-03-26 Cgi Pharmaceuticals, Inc. Substituted amides, methods of making, use thereof for the treatment of diseases such as cancer
WO2009051822A1 (en) 2007-10-19 2009-04-23 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
US20090105209A1 (en) 2007-10-23 2009-04-23 Roche Palo Alto Llc BTK protein kinase inhibitors
US20090155256A1 (en) 2007-10-17 2009-06-18 Wyeth Immunotherapy Regimes Dependent On APOE Status
WO2009077334A1 (en) 2007-12-14 2009-06-25 F. Hoffmann-La Roche Ag Novel imidazo[1,2-a]pyridine and imidazo[1,2-b]pyridazine derivatives
US7563869B2 (en) 2003-01-23 2009-07-21 Ono Pharmaceutical Co., Ltd. Substance specific to human PD-1
WO2009098144A1 (en) 2008-02-05 2009-08-13 F. Hoffmann-La Roche Ag Novel pyridinones and pyridazinones
US7595048B2 (en) 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
US7597889B1 (en) 1998-05-08 2009-10-06 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US7608429B2 (en) 2002-10-31 2009-10-27 Genentech, Inc. Methods and compositions for increasing antibody production
US20090318441A1 (en) 2006-05-26 2009-12-24 Astex Therapeutics Ltd. Pyrrolopyrimidine Compounds and Their Uses
WO2009158571A1 (en) 2008-06-27 2009-12-30 Avila Therapeutics And Uses Thereof Heteroaryl compounds and uses thereof
US20100004231A1 (en) 2008-07-02 2010-01-07 Roche Palo Alto Llc Inhibitors of burton's tyrosine kinase
WO2010006947A1 (en) 2008-07-15 2010-01-21 F. Hoffmann-La Roche Ag Novel phenyl-imidazopyridines and pyridazines
WO2010006970A1 (en) 2008-07-18 2010-01-21 F. Hoffmann-La Roche Ag Novel phenylimidazopyrazines
US20100016296A1 (en) 2007-10-19 2010-01-21 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
US7670600B2 (en) 2000-12-12 2010-03-02 MedImmine, LLC Molecules with extended half-lives, compositions and uses thereof
WO2010028236A1 (en) 2008-09-05 2010-03-11 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
US20100087464A1 (en) 2006-10-06 2010-04-08 Irm Llc Protein kinase inhibitors and methods for using thereof
US20100105676A1 (en) 2007-03-21 2010-04-29 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
WO2010051549A1 (en) 2008-10-31 2010-05-06 Genentech, Inc. Pyrazolopyrimidine jak inhibitor compounds and methods
US7718662B1 (en) 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
US20100144705A1 (en) 2008-12-05 2010-06-10 Principia Biopharma Inc. Egfr kinase knockdown via electrophilically enhanced inhibitors
WO2010068810A2 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
WO2010068788A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Heterocyclic amides as btk inhibitors
US20100151492A1 (en) 2008-11-28 2010-06-17 Emory University And Dana Farber Cancer Institute, Inc. Methods for the treatment of infections and tumors
WO2010068806A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Amide derivatives as btk inhibitors in the treatment of allergic, autoimmune and inflammatory disorders as well as cancer
US20100160303A1 (en) 2008-12-19 2010-06-24 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
US20100160292A1 (en) 2006-09-11 2010-06-24 Cgi Pharmaceuticals, Inc Kinase Inhibitors, and Methods of Using and Identifying Kinase Inhibitors
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
US20100197924A1 (en) 2008-12-22 2010-08-05 Millennium Pharmaceuticals, Inc. Preparation of aminotetralin compounds
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
US20100222325A1 (en) 2009-03-02 2010-09-02 Steven Berthel Inhibitors of Bruton's Tyrosine Kinase
US20100249092A1 (en) 2008-06-27 2010-09-30 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
WO2010122038A1 (en) 2009-04-24 2010-10-28 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
US7851598B2 (en) 2004-01-09 2010-12-14 Isis Innovation Limited Receptor modulators
US7863419B2 (en) 2003-08-22 2011-01-04 Biogen Idec Ma Inc. Antibodies having altered effector function and methods for making the same
US20110008369A1 (en) 2008-03-12 2011-01-13 Finnefrock Adam C Pd-1 binding proteins
WO2011006074A1 (en) 2009-07-09 2011-01-13 Array Biopharma Inc. SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS TRK KINASE INHIBITORS
US20110054151A1 (en) 2009-09-02 2011-03-03 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US20110118233A1 (en) 2008-05-06 2011-05-19 Cgi Pharmaceuticals, Inc. Certain Substituted Amides, Method of Making, and Method of Use Thereof
US20110124640A1 (en) 2008-07-24 2011-05-26 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
US20110159023A1 (en) 2008-08-25 2011-06-30 Solomon Langermann Pd-1 antagonists and methods for treating infectious disease
US20110171215A1 (en) 2008-09-12 2011-07-14 Isis Innovation Limited Pd-1 specific antibodies and uses thereof
US20110171220A1 (en) 2008-09-12 2011-07-14 Isis Innovation Limited Pd-1 specific antibodies and uses thereof
US20110177088A1 (en) 2007-08-17 2011-07-21 Universite De La Mediterranee Method for Treating and Diagnosing Hematologic Malignancies
US20110224235A1 (en) 2008-07-16 2011-09-15 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2011140488A1 (en) 2010-05-07 2011-11-10 Gilead Connecticut, Inc. Pyridone and aza-pyridone compounds and methods of use
US20110287032A1 (en) 2010-02-19 2011-11-24 Xencor, Inc. Novel ctla4-ig immunoadhesins
US20110301145A1 (en) 2008-11-12 2011-12-08 Gilead Connecticut INc. Pyridazinones, method of making, and method of use thereof
WO2011153514A2 (en) 2010-06-03 2011-12-08 Pharmacyclics, Inc. The use of inhibitors of bruton's tyrosine kinase (btk)
US20120028981A1 (en) 2008-11-05 2012-02-02 Principia Biopharma Inc. Kinase Knockdown Via Electrophilically Enhanced Inhibitors
US20120040961A1 (en) 2009-01-06 2012-02-16 Dana-Farber Cancer Institute Pyrimido-diazepinone kinase scaffold compounds and methods of treating disorders
WO2012020008A1 (en) 2010-08-12 2012-02-16 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
US20120053189A1 (en) 2010-06-28 2012-03-01 Pharmacyclics, Inc. Btk inhibitors for the treatment of immune mediated conditions
US20120076726A1 (en) 2006-03-23 2012-03-29 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US20120077832A1 (en) 2010-08-10 2012-03-29 Avila Therapeutics, Inc. Besylate salt of a btk inhibitor
US20120082702A1 (en) 2009-06-12 2012-04-05 Bristol-Meyers Squibb Company Nicotinamide compounds useful as kinase modulators
US20120129852A1 (en) 2009-08-11 2012-05-24 Bristol-Myers Squibb Company Azaindazoles as kinase inhibitors and use thereof
US20120157442A1 (en) 2009-09-04 2012-06-21 Sunesis Pharmaceuticals, Inc. Heteroaryl btk inhibitors
US20120157443A1 (en) 2009-09-04 2012-06-21 Sunesis Pharmaceuticals, Inc. Bruton's tyrosine kinase inhibitors
WO2012083370A1 (en) 2010-12-22 2012-06-28 Cephalon Australia Pty Ltd Modified antibody with improved half-life
US20120232054A1 (en) 2009-04-29 2012-09-13 Locus Pharmaceuticals, Inc. Pyrrolotriazine compounds
US20120237522A1 (en) 2008-10-02 2012-09-20 Seoul National University Industry Foundation Anticancer agent comprising anti-pd-1 antibody or anti-pd-l1 antibody
US20120251531A1 (en) 2011-03-29 2012-10-04 Genentech, Inc. ANTIBODY Fc VARIANTS
WO2012135408A1 (en) 2011-03-31 2012-10-04 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
WO2012135801A1 (en) 2011-04-01 2012-10-04 University Of Utah Research Foundation Substituted n-(3-(pyrimidin-4-yl)phenyl)acrylamide analogs as tyrosine receptor kinase btk inhibitors
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2012143522A1 (en) 2011-04-20 2012-10-26 Glaxo Group Limited Tetrahydropyrazolo [1,5 -a] pyrimidine as anti -tuberculosis compounds
WO2012158795A1 (en) 2011-05-17 2012-11-22 Principia Biopharma Inc. Pyrazolopyrimidine derivatives as tyrosine kinase inhibitors
WO2012156334A1 (en) 2011-05-17 2012-11-22 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
WO2012175692A1 (en) 2011-06-22 2012-12-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-axl antibodies and uses thereof
US20130004514A1 (en) 2011-06-06 2013-01-03 Novo Nordisk A/S Therapeutic antibodies
US20130079327A1 (en) 2010-05-31 2013-03-28 Shingo Yamamoto Purinone derivative
US20130089541A1 (en) 2010-03-29 2013-04-11 Zymeworks Inc Antibodies with Enhanced or Suppressed Effector Function
US20130096118A1 (en) 2010-06-16 2013-04-18 Chunjian Liu Carboline carboxamide compounds useful as kinase inhibitors
US20130116213A1 (en) 2010-06-23 2013-05-09 Hanmi Science Co., Ltd. Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
US20130259868A1 (en) 2010-07-15 2013-10-03 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
US20130261103A1 (en) 2010-09-01 2013-10-03 Genentech, Inc. Pyridazinones, method of making, and method of use thereof
US20130281432A1 (en) 2010-09-01 2013-10-24 Genentech, Inc. Pyridinones/pyrazinones, method of making, and method of use thereof
WO2013173223A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2013181634A2 (en) 2012-05-31 2013-12-05 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
US20140045833A1 (en) 2011-04-04 2014-02-13 Pharmascience Inc. Protein Kinase Inhibitors
US20140094459A1 (en) 2011-05-17 2014-04-03 Princpia Biopharma Inc. Tyrosine Kinase Inhibitors
WO2014055897A2 (en) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Human monoclonal anti-pd-l1 antibodies and methods of use
US20140107151A1 (en) 2011-05-17 2014-04-17 Principia Biophama Inc. Tyrosine kinase inhibitors
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
US20140162316A1 (en) 2009-10-28 2014-06-12 Janssen Biotech, Inc. Anti-GLP-1R Antibodies and Their Uses
US20140162983A1 (en) 2011-06-10 2014-06-12 Merck Patent Gmbh Compositions and Methods for the Production of Pyrimidine and Pyridine Compounds with BTK Inhibitory Activity
WO2014100079A1 (en) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
US20140243504A1 (en) 2013-02-01 2014-08-28 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US20140245468A1 (en) 2013-02-20 2014-08-28 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
US20140243306A1 (en) 2011-07-08 2014-08-28 Novartis Ag Novel pyrrolo pyrimidine derivatives
US20140271642A1 (en) 2013-03-15 2014-09-18 Regeneron Pharmaceuticals, Inc. Il-33 antagonists and uses thereof
WO2014173289A1 (en) 2013-04-25 2014-10-30 Beigene, Ltd. Fused heterocyclic compounds as protein kinase inhibitors
US20140341902A1 (en) 2011-08-01 2014-11-20 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US20140356363A1 (en) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Antigen Binding Proteins that Bind PD-1
US8911726B2 (en) 2004-09-22 2014-12-16 Kyowa Hakko Kirin Co., Ltd Stabilized human Igg4 antibodies
US20140377269A1 (en) 2012-12-19 2014-12-25 Adimab, Llc Multivalent antibody analogs, and methods of their preparation and use
WO2015061752A1 (en) 2013-10-25 2015-04-30 Pharmacyclics, Inc. Treatment using bruton's tyrosine kinase inhibitors and immunotherapy
US20150125444A1 (en) 2012-04-30 2015-05-07 Medimmune, Llc Molecules with Reduced Effector Function and Extended Half-Lives, Compositions, and Uses Thereof
US20150210763A1 (en) 2012-02-09 2015-07-30 Chugai Seiyaku Kabushiki Kaisha MODIFIED Fc REGION OF ANTIBODY
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
US9139653B1 (en) 2015-04-30 2015-09-22 Kymab Limited Anti-human OX40L antibodies and methods of treatment
US20150337053A1 (en) 2009-11-30 2015-11-26 Janssen Biotech, Inc. Antibody Fc Mutants with Ablated Effector Functions
US20150353631A1 (en) 2012-07-03 2015-12-10 Janssen Alzheimer Immunotherapy C-terminal and central epitope a-beta antibodies
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016008411A1 (en) 2014-07-18 2016-01-21 Beigene, Ltd. 5-amino-4-carbamoyl-pyrazole compounds as selective and irreversible t790m over wt-egfr kinase inhibitors and use thereof
WO2016025720A1 (en) 2014-08-14 2016-02-18 Assia Chemical Industries Ltd. Solid state forms of ibrutinib
WO2016024228A1 (en) 2014-08-11 2016-02-18 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor, a pi3k inhibitor, a jak-2 inhibitor, a pd-1 inhibitor and/or a pd-l1 inhibitor
WO2016064649A1 (en) 2014-10-24 2016-04-28 Eli Lilly And Company Therapy for urothelial carcinoma
WO2016087994A1 (en) 2014-12-05 2016-06-09 Acerta Pharma B.V. Btk inhibitors to treat solid tumors through modulation of the tumor microenvironment
WO2016100914A1 (en) 2014-12-18 2016-06-23 Gourlay Steven Treatment of pemphigus
WO2016105582A1 (en) 2014-12-24 2016-06-30 Nunn Philip A Compositions for ileo-jejunal drug delivery
US20170044260A1 (en) 2015-08-10 2017-02-16 Innovent Biologics, Inc. PD-1 Antibodies
WO2017046746A1 (en) 2015-09-15 2017-03-23 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor and a gitr binding molecule, a 4-1bb agonist, or an ox40 agonist
WO2017059224A2 (en) 2015-10-01 2017-04-06 Gilead Sciences, Inc. Combination of a btk inhibitor and a checkpoint inhibitor for treating cancers
US20180037655A1 (en) 2015-05-29 2018-02-08 Genentech, Inc. Therapeutic and diagnostic methods for cancer
WO2018033135A1 (en) 2016-08-19 2018-02-22 Beigene, Ltd. Use of a combination comprising a btk inhibitor for treating cancers
WO2018033853A2 (en) 2016-08-16 2018-02-22 Beigene, Ltd. Crystalline form of (s)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018137681A1 (en) 2017-01-25 2018-08-02 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a] pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018193105A1 (en) 2017-04-20 2018-10-25 Adc Therapeutics Sa Combination therapy
WO2019001417A1 (en) 2017-06-26 2019-01-03 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
WO2019034009A1 (en) 2017-08-12 2019-02-21 Beigene, Ltd. Btk INHIBITORS WITH IMPROVED DUAL SELECTIVITY
WO2019108795A1 (en) 2017-11-29 2019-06-06 Beigene Switzerland Gmbh Treatment of indolent or aggressive b-cell lymphomas using a combination comprising btk inhibitors
WO2019157353A1 (en) 2018-02-09 2019-08-15 Beigene, Ltd. Immunotherapy for urothelial carcinoma
US20200030339A1 (en) 2016-09-27 2020-01-30 Beigene, Ltd. Treatment cancers using a combination comprising parp inhibitors
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
US20200069666A1 (en) 2016-07-05 2020-03-05 Beigene, Ltd. Combination of a pd-1 antagonist and a raf inhibitor for treating cancer
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10858435B2 (en) 2018-03-08 2020-12-08 Ultrahuman Nine Limited PD1 binding agents

Patent Citations (313)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1412017A (en) 1971-12-09 1975-10-29 Icn Pharmaceuticals Pyrazolo 1,5a pyrimidines
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US6632927B2 (en) 1989-12-21 2003-10-14 Celltech Therapeutics Limited Humanized antibodies
US5994514A (en) 1991-08-14 1999-11-30 Genentech, Inc. Immunoglobulin variants
US6165745A (en) 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5629204A (en) 1994-03-01 1997-05-13 Ono Pharmaceutical Co., Ltd. Peptide related to human programmed cell death and DNA encoding it
US5698520A (en) 1994-03-01 1997-12-16 Ono Pharmaceutical Co., Ltd. Peptide related to human programmed cell death and DNA encoding the same
JPH07278148A (en) 1994-11-18 1995-10-24 Sankyo Co Ltd Imidazopyrazole derivative
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US7297775B2 (en) 1998-04-02 2007-11-20 Genentech, Inc. Polypeptide variants
US7741072B2 (en) 1998-04-02 2010-06-22 Genentech, Inc. Polypeptide variants
US7364731B2 (en) 1998-04-02 2008-04-29 Genentech, Inc. Polypeptide variants
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US7597889B1 (en) 1998-05-08 2009-10-06 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
US7335742B2 (en) 1999-01-15 2008-02-26 Genentech, Inc. Polypeptide variants with altered effector function
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US7122637B2 (en) 1999-01-15 2006-10-17 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7790858B2 (en) 1999-01-15 2010-09-07 Genentech, Inc. Polypeptide variants with altered effector function
US6808710B1 (en) 1999-08-23 2004-10-26 Genetics Institute, Inc. Downmodulating an immune response with multivalent antibodies to PD-1
US7038013B2 (en) 1999-08-23 2006-05-02 Dana-Faber Cancer Institute, Inc. B7-4 polypeptides and uses therefor
US7101550B2 (en) 1999-08-23 2006-09-05 Dana-Farber Cancer Institute, Inc. PD-1, a receptor for B7-4, and uses therefor
US7635757B2 (en) 1999-08-23 2009-12-22 Dana-Farber Cancer Institute, Inc. B7-4 Antibodies and uses therefor
US7638492B2 (en) 1999-08-23 2009-12-29 Dana-Farber Cancer Institute, Inc. Methods of upmodulating an immune response with non-activating forms of B7-4
US6936704B1 (en) 1999-08-23 2005-08-30 Dana-Farber Cancer Institute, Inc. Nucleic acids encoding costimulatory molecule B7-4
WO2001016138A1 (en) 1999-08-27 2001-03-08 Abbott Laboratories Sulfonylphenylpyrazole compounds useful as cox-2 inhibitors
WO2001019829A2 (en) 1999-09-17 2001-03-22 Basf Aktiengesellschaft Pyrazolopyrimidines as therapeutic agents
US20040038339A1 (en) 2000-03-24 2004-02-26 Peter Kufer Multifunctional polypeptides comprising a binding site to an epitope of the nkg2d receptor complex
US7416726B2 (en) 2000-04-13 2008-08-26 The Rockefeller University Enhancement of antibody-mediated immune responses
US20110052584A1 (en) 2000-04-13 2011-03-03 The Rockefeller University Method of enhancement of cytotoxicity in antibody mediated immune responses
US20020094989A1 (en) 2000-10-11 2002-07-18 Hale Jeffrey J. Pyrrolidine modulators of CCR5 chemokine receptor activity
US7414171B2 (en) 2000-11-15 2008-08-19 Ono Pharmaceutical Co., Ltd. PD-1-lacking mouse and use thereof
US7670600B2 (en) 2000-12-12 2010-03-02 MedImmine, LLC Molecules with extended half-lives, compositions and uses thereof
WO2002050071A1 (en) 2000-12-21 2002-06-27 Bristol-Myers Squibb Company Thiazolyl inhibitors of tec family tyrosine kinases
US20050202028A1 (en) 2001-01-17 2005-09-15 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20060263856A1 (en) 2001-03-07 2006-11-23 Emd Lexigen Research Center Corp. Expression technology for proteins containing a hybrid isotype antibody moiety
WO2002072576A1 (en) 2001-03-09 2002-09-19 Pfizer Products Inc. Benzimidazole anti-inflammatory compounds
US7029674B2 (en) 2001-04-02 2006-04-18 Wyeth Methods for downmodulating immune cells using an antibody to PD-1
WO2003004497A1 (en) 2001-07-05 2003-01-16 Sumitomo Pharmaceuticals Company, Limited Novel heterocyclic compound
US20070160597A1 (en) 2002-03-01 2007-07-12 Xencor, Inc. Optimized Fc variants and methods for their generation
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20090068175A1 (en) 2002-03-01 2009-03-12 Xencor, Inc. Optimized FC Variants and Methods for Their Generation
US9073994B2 (en) 2002-07-03 2015-07-07 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
US7595048B2 (en) 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
US9067999B1 (en) 2002-07-03 2015-06-30 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US20140314714A1 (en) 2002-07-03 2014-10-23 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US8728474B2 (en) 2002-07-03 2014-05-20 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
JP2006510582A (en) 2002-08-26 2006-03-30 武田薬品工業株式会社 Calcium receptor modulating compound and use thereof
CN1771231A (en) 2002-08-26 2006-05-10 武田药品工业株式会社 Calcium receptor modulating compound and use thereof
WO2004017908A2 (en) 2002-08-26 2004-03-04 Takeda Pharmaceutical Company Limited Calcium receptor modulating compound and use thereof
US7655783B2 (en) 2002-10-31 2010-02-02 Genentech, Inc. Methods and compositions for increasing antibody production
US7608429B2 (en) 2002-10-31 2009-10-27 Genentech, Inc. Methods and compositions for increasing antibody production
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
CN101899114A (en) 2002-12-23 2010-12-01 惠氏公司 Anti-PD-1 antibody and uses thereof
CN1753912A (en) 2002-12-23 2006-03-29 惠氏公司 Antibodies against PD-1 and uses therefor
US8088905B2 (en) 2002-12-23 2012-01-03 Wyeth Nucleic acids encoding antibodies against PD-1
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20050064514A1 (en) 2003-01-09 2005-03-24 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US7563869B2 (en) 2003-01-23 2009-07-21 Ono Pharmaceutical Co., Ltd. Substance specific to human PD-1
US7998479B2 (en) 2003-01-23 2011-08-16 Ono Pharmaceutical Co., Ltd. Substance specific to human PD-1
US8246955B2 (en) 2003-01-23 2012-08-21 Ono Pharmaceutical Co., Ltd. Substance specific to human PD-1
US20060121032A1 (en) 2003-03-03 2006-06-08 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
WO2005014599A1 (en) 2003-06-04 2005-02-17 Cellular Genomics, Inc. Imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of bruton’s tyrosine kinase by such compounds
US20060183746A1 (en) 2003-06-04 2006-08-17 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of Bruton's tyrosine kinase by such compounds
US7393848B2 (en) 2003-06-30 2008-07-01 Cgi Pharmaceuticals, Inc. Certain heterocyclic substituted imidazo[1,2-A]pyrazin-8-ylamines and methods of inhibition of Bruton's tyrosine kinase by such compounds
WO2005005429A1 (en) 2003-06-30 2005-01-20 Cellular Genomics, Inc. Certain heterocyclic substituted imidazo[1,2-a]pyrazin-8-ylamines and methods of inhibition of bruton’s tyrosine kinase by such compounds
WO2005011597A2 (en) 2003-07-29 2005-02-10 Irm Llc Compounds and compositions as protein kinase inhibitors
US7863419B2 (en) 2003-08-22 2011-01-04 Biogen Idec Ma Inc. Antibodies having altered effector function and methods for making the same
WO2005047290A2 (en) 2003-11-11 2005-05-26 Cellular Genomics Inc. Imidazo[1,2-a] pyrazin-8-ylamines as kinase inhibitors
WO2005077981A2 (en) 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES
US8945561B2 (en) 2004-01-09 2015-02-03 Isis Innovation Limited Receptor modulators
US7851598B2 (en) 2004-01-09 2010-12-14 Isis Innovation Limited Receptor modulators
US8911726B2 (en) 2004-09-22 2014-12-16 Kyowa Hakko Kirin Co., Ltd Stabilized human Igg4 antibodies
US20100317834A1 (en) 2004-10-21 2010-12-16 Xencor, Inc. IgG Immunoglobulin Variants with Optimized Effector Function
US20060134105A1 (en) 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
WO2006053121A2 (en) 2004-11-10 2006-05-18 Cgi Pharmaceuticals, Inc. Imidazo[1 , 2-a] pyrazin-8-ylamines useful as modulators of kinase activity
US20060178367A1 (en) 2004-11-10 2006-08-10 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines, method of making, and method of use thereof
WO2006065946A1 (en) 2004-12-16 2006-06-22 Vertex Pharmaceuticals Incorporated Pyrid-2-ones useful as inhibitors of tec family protein kinases for the treatment of inflammatory, proliferative and immunologically-mediated diseases
WO2006084015A2 (en) 2005-02-04 2006-08-10 Genentech, Inc. Raf inhibitor compounds and methods
WO2006099075A2 (en) 2005-03-10 2006-09-21 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
US9084776B2 (en) 2005-05-09 2015-07-21 E.R. Squibb & Sons, L.L.C. Methods for treating cancer using anti-PD-1 antibodies
CN101213297A (en) 2005-05-09 2008-07-02 小野药品工业株式会社 Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
US9492540B2 (en) 2005-05-09 2016-11-15 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies
US8779105B2 (en) 2005-05-09 2014-07-15 Medarex, L.L.C. Monoclonal antibodies to programmed death 1 (PD-1)
JP2009155338A (en) 2005-05-09 2009-07-16 Ono Pharmaceut Co Ltd Cancer therapeutic method singly using human monoclonal antibody to programmed death 1 (pd-1) and anti-pd-1 antibody or concurrently using with other immunotherapy
KR20080011428A (en) 2005-05-09 2008-02-04 오노 야꾸힝 고교 가부시키가이샤 Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
US20090217401A1 (en) 2005-05-09 2009-08-27 Medarex, Inc Human Monoclonal Antibodies To Programmed Death 1(PD-1) And Methods For Treating Cancer Using Anti-PD-1 Antibodies Alone or in Combination with Other Immunotherapeutics
WO2006133396A2 (en) 2005-06-08 2006-12-14 Dana-Farber Cancer Institute Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
CN101355965A (en) 2005-06-08 2009-01-28 达纳-法伯癌症研究院 Methods and compositions for the treatment of persistent infections and cancers by inhibiting the programmed cell death 1 (PD-1) phthway
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
JP2008544755A (en) 2005-07-01 2008-12-11 メダレックス インコーポレーティッド Human monoclonal antibody against programmed death ligand 1 (PD-L1)
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007027729A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3, 5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
WO2007027594A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
WO2007026720A1 (en) 2005-08-31 2007-03-08 Taisho Pharmaceutical Co., Ltd. Ring-fused pyrazole derivative
WO2007026950A1 (en) 2005-09-01 2007-03-08 Astellas Pharma Inc. Pyridazinone derivatives used for the treatment of pain
WO2007067444A1 (en) 2005-12-08 2007-06-14 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
US20100035841A1 (en) 2006-01-13 2010-02-11 Pharmacyclics, Inc. Inhibitors of tyrosine kinases and uses thereof
WO2007087068A2 (en) 2006-01-13 2007-08-02 Pharmacyclics, Inc. Inhibitors of tyrosine kinases and uses thereof
US20120076726A1 (en) 2006-03-23 2012-03-29 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
WO2007136572A2 (en) 2006-05-15 2007-11-29 Merck & Co., Inc. Antidiabetic bicyclic compounds
WO2007136790A2 (en) 2006-05-18 2007-11-29 Mannkind Corporation Intracellular kinase inhibitors
US20090318441A1 (en) 2006-05-26 2009-12-24 Astex Therapeutics Ltd. Pyrrolopyrimidine Compounds and Their Uses
CN101104640A (en) 2006-07-10 2008-01-16 苏州大学 Preparation for anti human PD-L1 monoclonal antibody and application thereof
US20100160292A1 (en) 2006-09-11 2010-06-24 Cgi Pharmaceuticals, Inc Kinase Inhibitors, and Methods of Using and Identifying Kinase Inhibitors
WO2008033854A1 (en) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
WO2008033857A2 (en) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Substituted amides, method of making, and method of use thereof
WO2008033834A1 (en) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Pyrimidines derivatives and their use as kinase inhibitors
US20100254905A1 (en) 2006-09-22 2010-10-07 Lee Honigberg Inhibitors of bruton's tyrosine kinase
WO2008039218A2 (en) 2006-09-22 2008-04-03 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US20080076921A1 (en) 2006-09-22 2008-03-27 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US7514444B2 (en) 2006-09-22 2009-04-07 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
JP2010504324A (en) 2006-09-22 2010-02-12 ファーマサイクリクス,インコーポレイテッド Bruton tyrosine kinase inhibitor
US20080139582A1 (en) 2006-09-22 2008-06-12 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US20100087464A1 (en) 2006-10-06 2010-04-08 Irm Llc Protein kinase inhibitors and methods for using thereof
WO2008054827A2 (en) 2006-11-03 2008-05-08 Pharmacyclics, Inc. Bruton's tyrosine kinase activity probe and method of using
US20100105676A1 (en) 2007-03-21 2010-04-29 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
WO2008144253A1 (en) 2007-05-14 2008-11-27 Irm Llc Protein kinase inhibitors and methods for using thereof
WO2008145142A1 (en) 2007-05-31 2008-12-04 Genmab A/S Stable igg4 antibodies
JP2010528993A (en) 2007-05-31 2010-08-26 ゲンマブ エー/エス Stable IgG4 antibody
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
JP2012254092A (en) 2007-06-18 2012-12-27 Msd Oss Bv Antibody to human programmed death receptor pd-1
KR20100054780A (en) 2007-06-18 2010-05-25 엔.브이.오가논 Antibodies to human programmed death receptor pd-1
US20110177088A1 (en) 2007-08-17 2011-07-21 Universite De La Mediterranee Method for Treating and Diagnosing Hematologic Malignancies
WO2009039397A2 (en) 2007-09-20 2009-03-26 Cgi Pharmaceuticals, Inc. Substituted amides, methods of making, use thereof for the treatment of diseases such as cancer
US20090155256A1 (en) 2007-10-17 2009-06-18 Wyeth Immunotherapy Regimes Dependent On APOE Status
US20100016296A1 (en) 2007-10-19 2010-01-21 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
WO2009051822A1 (en) 2007-10-19 2009-04-23 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
US20090105209A1 (en) 2007-10-23 2009-04-23 Roche Palo Alto Llc BTK protein kinase inhibitors
WO2009077334A1 (en) 2007-12-14 2009-06-25 F. Hoffmann-La Roche Ag Novel imidazo[1,2-a]pyridine and imidazo[1,2-b]pyridazine derivatives
WO2009098144A1 (en) 2008-02-05 2009-08-13 F. Hoffmann-La Roche Ag Novel pyridinones and pyridazinones
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US20110008369A1 (en) 2008-03-12 2011-01-13 Finnefrock Adam C Pd-1 binding proteins
US20110118233A1 (en) 2008-05-06 2011-05-19 Cgi Pharmaceuticals, Inc. Certain Substituted Amides, Method of Making, and Method of Use Thereof
WO2009158571A1 (en) 2008-06-27 2009-12-30 Avila Therapeutics And Uses Thereof Heteroaryl compounds and uses thereof
US20100029610A1 (en) 2008-06-27 2010-02-04 Avila Therapeutics, Inc. Heteroaryl Compounds and Uses Thereof
US20100249092A1 (en) 2008-06-27 2010-09-30 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
US20100004231A1 (en) 2008-07-02 2010-01-07 Roche Palo Alto Llc Inhibitors of burton's tyrosine kinase
WO2010000633A1 (en) 2008-07-02 2010-01-07 F. Hoffmann-La Roche Ag Novel phenylpyrazinones as kinase inhibitors
US20100016301A1 (en) 2008-07-15 2010-01-21 Roche Palo Alto Llc Inhibitors of bruton's tyrosine kinase
WO2010006947A1 (en) 2008-07-15 2010-01-21 F. Hoffmann-La Roche Ag Novel phenyl-imidazopyridines and pyridazines
US20110224235A1 (en) 2008-07-16 2011-09-15 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors
WO2010006970A1 (en) 2008-07-18 2010-01-21 F. Hoffmann-La Roche Ag Novel phenylimidazopyrazines
US20110124640A1 (en) 2008-07-24 2011-05-26 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
US20110159023A1 (en) 2008-08-25 2011-06-30 Solomon Langermann Pd-1 antagonists and methods for treating infectious disease
US20110195068A1 (en) 2008-08-25 2011-08-11 Solomon Langermann Pd-1 antagonists and methods of use thereof
WO2010028236A1 (en) 2008-09-05 2010-03-11 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
US20110171215A1 (en) 2008-09-12 2011-07-14 Isis Innovation Limited Pd-1 specific antibodies and uses thereof
US20110171220A1 (en) 2008-09-12 2011-07-14 Isis Innovation Limited Pd-1 specific antibodies and uses thereof
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
CN102264762A (en) 2008-09-26 2011-11-30 达纳-法伯癌症研究公司 Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US20120237522A1 (en) 2008-10-02 2012-09-20 Seoul National University Industry Foundation Anticancer agent comprising anti-pd-1 antibody or anti-pd-l1 antibody
US8617546B2 (en) 2008-10-02 2013-12-31 Seoul National University Industry Foundation Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody
WO2010051549A1 (en) 2008-10-31 2010-05-06 Genentech, Inc. Pyrazolopyrimidine jak inhibitor compounds and methods
US20120028981A1 (en) 2008-11-05 2012-02-02 Principia Biopharma Inc. Kinase Knockdown Via Electrophilically Enhanced Inhibitors
US20110301145A1 (en) 2008-11-12 2011-12-08 Gilead Connecticut INc. Pyridazinones, method of making, and method of use thereof
US20100151492A1 (en) 2008-11-28 2010-06-17 Emory University And Dana Farber Cancer Institute, Inc. Methods for the treatment of infections and tumors
WO2010065898A2 (en) 2008-12-05 2010-06-10 Principia Biopharma Inc. Egfr kinase knockdown via electrophilically enhanced inhibitors
US20100144705A1 (en) 2008-12-05 2010-06-10 Principia Biopharma Inc. Egfr kinase knockdown via electrophilically enhanced inhibitors
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
CN102245640A (en) 2008-12-09 2011-11-16 霍夫曼-拉罗奇有限公司 Anti-PD-L1 antibodies and their use to enhance T-cell function
JP2012511329A (en) 2008-12-09 2012-05-24 ジェネンテック, インコーポレイテッド Anti-PD-L1 antibodies and their use to enhance T cell function
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2010068806A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Amide derivatives as btk inhibitors in the treatment of allergic, autoimmune and inflammatory disorders as well as cancer
WO2010068788A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Heterocyclic amides as btk inhibitors
WO2010068810A2 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
US8084620B2 (en) 2008-12-19 2011-12-27 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
US20120058996A1 (en) 2008-12-19 2012-03-08 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
US20100160303A1 (en) 2008-12-19 2010-06-24 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
US20100197924A1 (en) 2008-12-22 2010-08-05 Millennium Pharmaceuticals, Inc. Preparation of aminotetralin compounds
US20120040961A1 (en) 2009-01-06 2012-02-16 Dana-Farber Cancer Institute Pyrimido-diazepinone kinase scaffold compounds and methods of treating disorders
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
US20100222325A1 (en) 2009-03-02 2010-09-02 Steven Berthel Inhibitors of Bruton's Tyrosine Kinase
WO2010122038A1 (en) 2009-04-24 2010-10-28 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
US20120232054A1 (en) 2009-04-29 2012-09-13 Locus Pharmaceuticals, Inc. Pyrrolotriazine compounds
US20120082702A1 (en) 2009-06-12 2012-04-05 Bristol-Meyers Squibb Company Nicotinamide compounds useful as kinase modulators
WO2011006074A1 (en) 2009-07-09 2011-01-13 Array Biopharma Inc. SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS TRK KINASE INHIBITORS
US20120129852A1 (en) 2009-08-11 2012-05-24 Bristol-Myers Squibb Company Azaindazoles as kinase inhibitors and use thereof
US20110054151A1 (en) 2009-09-02 2011-03-03 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US20120157442A1 (en) 2009-09-04 2012-06-21 Sunesis Pharmaceuticals, Inc. Heteroaryl btk inhibitors
US20120157443A1 (en) 2009-09-04 2012-06-21 Sunesis Pharmaceuticals, Inc. Bruton's tyrosine kinase inhibitors
US7718662B1 (en) 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
CN102656173A (en) 2009-10-12 2012-09-05 药品循环公司 Inhibitors of Bruton's tyrosine kinase
US20140162316A1 (en) 2009-10-28 2014-06-12 Janssen Biotech, Inc. Anti-GLP-1R Antibodies and Their Uses
US20150337053A1 (en) 2009-11-30 2015-11-26 Janssen Biotech, Inc. Antibody Fc Mutants with Ablated Effector Functions
US20110287032A1 (en) 2010-02-19 2011-11-24 Xencor, Inc. Novel ctla4-ig immunoadhesins
US20130089541A1 (en) 2010-03-29 2013-04-11 Zymeworks Inc Antibodies with Enhanced or Suppressed Effector Function
WO2011140488A1 (en) 2010-05-07 2011-11-10 Gilead Connecticut, Inc. Pyridone and aza-pyridone compounds and methods of use
US20130079327A1 (en) 2010-05-31 2013-03-28 Shingo Yamamoto Purinone derivative
WO2011153514A2 (en) 2010-06-03 2011-12-08 Pharmacyclics, Inc. The use of inhibitors of bruton's tyrosine kinase (btk)
US20130096118A1 (en) 2010-06-16 2013-04-18 Chunjian Liu Carboline carboxamide compounds useful as kinase inhibitors
US20130116213A1 (en) 2010-06-23 2013-05-09 Hanmi Science Co., Ltd. Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
US20120053189A1 (en) 2010-06-28 2012-03-01 Pharmacyclics, Inc. Btk inhibitors for the treatment of immune mediated conditions
US20130259868A1 (en) 2010-07-15 2013-10-03 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
US20120077832A1 (en) 2010-08-10 2012-03-29 Avila Therapeutics, Inc. Besylate salt of a btk inhibitor
WO2012020008A1 (en) 2010-08-12 2012-02-16 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
US20130281432A1 (en) 2010-09-01 2013-10-24 Genentech, Inc. Pyridinones/pyrazinones, method of making, and method of use thereof
US20130261103A1 (en) 2010-09-01 2013-10-03 Genentech, Inc. Pyridazinones, method of making, and method of use thereof
WO2012083370A1 (en) 2010-12-22 2012-06-28 Cephalon Australia Pty Ltd Modified antibody with improved half-life
US20120251531A1 (en) 2011-03-29 2012-10-04 Genentech, Inc. ANTIBODY Fc VARIANTS
WO2012130831A1 (en) 2011-03-29 2012-10-04 Roche Glycart Ag Antibody fc variants
WO2012135408A1 (en) 2011-03-31 2012-10-04 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
WO2012135801A1 (en) 2011-04-01 2012-10-04 University Of Utah Research Foundation Substituted n-(3-(pyrimidin-4-yl)phenyl)acrylamide analogs as tyrosine receptor kinase btk inhibitors
US20140045833A1 (en) 2011-04-04 2014-02-13 Pharmascience Inc. Protein Kinase Inhibitors
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2012143522A1 (en) 2011-04-20 2012-10-26 Glaxo Group Limited Tetrahydropyrazolo [1,5 -a] pyrimidine as anti -tuberculosis compounds
US20140044738A1 (en) 2011-04-20 2014-02-13 Amplimmune, Inc. Antibodies And Other Molecules That Bind B7-H1 And PD-1
WO2012156334A1 (en) 2011-05-17 2012-11-22 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
US20140221398A1 (en) 2011-05-17 2014-08-07 Principia Biopharma Inc. Pyrazolopyrimidine derivatives as tyrosine kinase inhibitors
US20140094459A1 (en) 2011-05-17 2014-04-03 Princpia Biopharma Inc. Tyrosine Kinase Inhibitors
WO2012158795A1 (en) 2011-05-17 2012-11-22 Principia Biopharma Inc. Pyrazolopyrimidine derivatives as tyrosine kinase inhibitors
US20140107151A1 (en) 2011-05-17 2014-04-17 Principia Biophama Inc. Tyrosine kinase inhibitors
US20130004514A1 (en) 2011-06-06 2013-01-03 Novo Nordisk A/S Therapeutic antibodies
US20150044231A1 (en) 2011-06-06 2015-02-12 Novo Nordisk A/S Therapeutic Antibodies
US20140162983A1 (en) 2011-06-10 2014-06-12 Merck Patent Gmbh Compositions and Methods for the Production of Pyrimidine and Pyridine Compounds with BTK Inhibitory Activity
WO2012175692A1 (en) 2011-06-22 2012-12-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-axl antibodies and uses thereof
US20140243306A1 (en) 2011-07-08 2014-08-28 Novartis Ag Novel pyrrolo pyrimidine derivatives
US20140341902A1 (en) 2011-08-01 2014-11-20 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US10487147B2 (en) 2011-11-28 2019-11-26 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US9624298B2 (en) 2011-11-28 2017-04-18 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
US20150210763A1 (en) 2012-02-09 2015-07-30 Chugai Seiyaku Kabushiki Kaisha MODIFIED Fc REGION OF ANTIBODY
US20150125444A1 (en) 2012-04-30 2015-05-07 Medimmune, Llc Molecules with Reduced Effector Function and Extended Half-Lives, Compositions, and Uses Thereof
WO2013173223A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
US20130309250A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
US10058609B2 (en) 2012-05-31 2018-08-28 Sorrento Therapeutics, Inc. Antigen binding proteins that bind PD-L1
WO2013181634A2 (en) 2012-05-31 2013-12-05 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
US9175082B2 (en) 2012-05-31 2015-11-03 Sorrento Therapeutics, Inc. Antigen binding proteins that bind PD-L1
US20150353631A1 (en) 2012-07-03 2015-12-10 Janssen Alzheimer Immunotherapy C-terminal and central epitope a-beta antibodies
WO2014055897A2 (en) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Human monoclonal anti-pd-l1 antibodies and methods of use
US20140377269A1 (en) 2012-12-19 2014-12-25 Adimab, Llc Multivalent antibody analogs, and methods of their preparation and use
WO2014100079A1 (en) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
US20140243504A1 (en) 2013-02-01 2014-08-28 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US20140245468A1 (en) 2013-02-20 2014-08-28 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
US20140271642A1 (en) 2013-03-15 2014-09-18 Regeneron Pharmaceuticals, Inc. Il-33 antagonists and uses thereof
US20150259354A1 (en) 2013-04-25 2015-09-17 Beigene, Ltd. Fused heterocyclic compounds as protein kinase inhibitors
WO2014173289A1 (en) 2013-04-25 2014-10-30 Beigene, Ltd. Fused heterocyclic compounds as protein kinase inhibitors
US10570139B2 (en) 2013-04-25 2020-02-25 Beigene Switzerland Gmbh Substituted pyrazolo[1,5-a]pyrimidines as Bruton's tyrosine kinase modulators
US10005782B2 (en) 2013-04-25 2018-06-26 Beigene, Ltd. Substituted pyrazolo[1,5-a]pyrimidines as bruton's tyrosine kinase modulators
CN104884458A (en) 2013-04-25 2015-09-02 百济神州有限公司 Fused heterocyclic compounds as protein kinase inhibitors
US20180251466A1 (en) 2013-04-25 2018-09-06 Beigene, Ltd. SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINES AS BRUTON'S TYROSINE KINASE MODULATORS
US20200148690A1 (en) 2013-04-25 2020-05-14 Beigene Switzerland Gmbh SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINES AS BRUTON'S TYROSINE KINASE MODULATORS
US20170073349A1 (en) 2013-04-25 2017-03-16 Beigene, Ltd. Fused heterocyclic compounds as protein kinase inhibitors
US9556188B2 (en) 2013-04-25 2017-01-31 Beigene, Ltd. Substituted imidazo[1,2-b]pyrazoles as bruton'S tyrosine kinase modulators
US9447106B2 (en) 2013-04-25 2016-09-20 Beigene, Ltd. Substituted pyrazolo[1,5-a]pyrimidines as bruton's tyrosine kinase modulators
US20160083392A1 (en) 2013-04-25 2016-03-24 Beigene, Ltd. Fused heterocyclic compounds as protein kinase inhibitors
US20140356363A1 (en) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Antigen Binding Proteins that Bind PD-1
US20150079109A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
US20200216535A1 (en) 2013-09-13 2020-07-09 Beigene Switzerland Gmbh Anti-pd1 antibodies and their use as therapeutics and diagnostics
US20210230274A1 (en) 2013-09-13 2021-07-29 Beigene Switzerland Gmbh Anti-pd1 antibodies and their use as therapeutics and diagnostics
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2015035606A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-pd1 antibodies and their use as therapeutics and diagnostics
CN105531288A (en) 2013-09-13 2016-04-27 百济神州有限公司 Anti-PD1 antibodies and their use as therapeutics and diagnostics
US10519235B2 (en) 2013-09-13 2019-12-31 Beigene Switzerland Gmbh Anti-PD1 antibodies and their use as therapeutics and diagnostics
US20180251551A1 (en) 2013-09-13 2018-09-06 Beigene, Ltd. Anti-pd1 antibodies and their use as therapeutics and diagnostics
US20150315274A1 (en) 2013-09-13 2015-11-05 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
US9988450B2 (en) 2013-09-13 2018-06-05 Beigene Switzerland Gmbh Anti-PD1 antibodies and their use as therapeutics and diagnostics
US9217034B2 (en) 2013-09-13 2015-12-22 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
US9834606B2 (en) 2013-09-13 2017-12-05 Beigene, Ltd Anti-PD1 antibodies and their use as therapeutics and diagnostics
CN107090041A (en) 2013-09-13 2017-08-25 百济神州有限公司 Anti- PD1 antibody and its purposes as therapeutic agent and diagnosticum
CN107011441A (en) 2013-09-13 2017-08-04 百济神州有限公司 Anti- PD1 antibody and its purposes as therapeutic agent and diagnosticum
WO2015061752A1 (en) 2013-10-25 2015-04-30 Pharmacyclics, Inc. Treatment using bruton's tyrosine kinase inhibitors and immunotherapy
CN106103485A (en) 2014-01-24 2016-11-09 达纳-法伯癌症研究公司 Antibody molecule of PD 1 and application thereof
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
US20200283527A1 (en) 2014-07-03 2020-09-10 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
US10544225B2 (en) 2014-07-03 2020-01-28 Beigene, Ltd. Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
US20180215825A1 (en) 2014-07-03 2018-08-02 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016008411A1 (en) 2014-07-18 2016-01-21 Beigene, Ltd. 5-amino-4-carbamoyl-pyrazole compounds as selective and irreversible t790m over wt-egfr kinase inhibitors and use thereof
WO2016024228A1 (en) 2014-08-11 2016-02-18 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor, a pi3k inhibitor, a jak-2 inhibitor, a pd-1 inhibitor and/or a pd-l1 inhibitor
WO2016025720A1 (en) 2014-08-14 2016-02-18 Assia Chemical Industries Ltd. Solid state forms of ibrutinib
WO2016064649A1 (en) 2014-10-24 2016-04-28 Eli Lilly And Company Therapy for urothelial carcinoma
WO2016087994A1 (en) 2014-12-05 2016-06-09 Acerta Pharma B.V. Btk inhibitors to treat solid tumors through modulation of the tumor microenvironment
WO2016100914A1 (en) 2014-12-18 2016-06-23 Gourlay Steven Treatment of pemphigus
WO2016105582A1 (en) 2014-12-24 2016-06-30 Nunn Philip A Compositions for ileo-jejunal drug delivery
US9139653B1 (en) 2015-04-30 2015-09-22 Kymab Limited Anti-human OX40L antibodies and methods of treatment
US20180037655A1 (en) 2015-05-29 2018-02-08 Genentech, Inc. Therapeutic and diagnostic methods for cancer
US20170044260A1 (en) 2015-08-10 2017-02-16 Innovent Biologics, Inc. PD-1 Antibodies
WO2017025016A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017046746A1 (en) 2015-09-15 2017-03-23 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor and a gitr binding molecule, a 4-1bb agonist, or an ox40 agonist
WO2017059224A2 (en) 2015-10-01 2017-04-06 Gilead Sciences, Inc. Combination of a btk inhibitor and a checkpoint inhibitor for treating cancers
US20210228553A1 (en) 2016-07-05 2021-07-29 Beigene, Ltd. Combination of a pd-1 antagonist and a raf inhibitor for treating cancer
US20200069666A1 (en) 2016-07-05 2020-03-05 Beigene, Ltd. Combination of a pd-1 antagonist and a raf inhibitor for treating cancer
US10864203B2 (en) 2016-07-05 2020-12-15 Beigene, Ltd. Combination of a PD-1 antagonist and a RAF inhibitor for treating cancer
US10927117B2 (en) 2016-08-16 2021-02-23 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US20210130363A1 (en) 2016-08-16 2021-05-06 Beigene Switzerland Gmbh CRYSTALLINE FORM OF (S)-7-(1-ACRYLOYLPIPERIDIN-4-YL)-2-(4-PHENOXYPHENYL)-4,5,6,7-TETRA-HYDROPYRAZOLO[1,5-a]PYRIMIDINE-3-CARBOXAMIDE, PREPARATION, AND USES THEREOF
US20190169201A1 (en) 2016-08-16 2019-06-06 Beigene, Ltd. Crystalline form of (s)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018033853A2 (en) 2016-08-16 2018-02-22 Beigene, Ltd. Crystalline form of (s)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US20210275530A1 (en) 2016-08-19 2021-09-09 Beigene, Ltd. Treatment cancers using a combination comprising btk inhibitors
WO2018033135A1 (en) 2016-08-19 2018-02-22 Beigene, Ltd. Use of a combination comprising a btk inhibitor for treating cancers
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US20200030339A1 (en) 2016-09-27 2020-01-30 Beigene, Ltd. Treatment cancers using a combination comprising parp inhibitors
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
WO2018137681A1 (en) 2017-01-25 2018-08-02 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a] pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018193105A1 (en) 2017-04-20 2018-10-25 Adc Therapeutics Sa Combination therapy
US20210147543A1 (en) 2017-06-26 2021-05-20 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
WO2019001417A1 (en) 2017-06-26 2019-01-03 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
US20200181150A1 (en) 2017-08-12 2020-06-11 Beigene, Ltd. Btk inhibitors with improved dual selectivity
WO2019034009A1 (en) 2017-08-12 2019-02-21 Beigene, Ltd. Btk INHIBITORS WITH IMPROVED DUAL SELECTIVITY
US20200368237A1 (en) 2017-11-29 2020-11-26 Beigene Switzerland Gmbh Treatment of indolent or aggressive b-cell lymphomas using a combination comprising btk inhibitors
WO2019108795A1 (en) 2017-11-29 2019-06-06 Beigene Switzerland Gmbh Treatment of indolent or aggressive b-cell lymphomas using a combination comprising btk inhibitors
US20210040213A1 (en) 2018-02-09 2021-02-11 Beigene, Ltd. Immunomonotherapy for urothelial carcinoma
WO2019157353A1 (en) 2018-02-09 2019-08-15 Beigene, Ltd. Immunotherapy for urothelial carcinoma
US10858435B2 (en) 2018-03-08 2020-12-08 Ultrahuman Nine Limited PD1 binding agents

Non-Patent Citations (112)

* Cited by examiner, † Cited by third party
Title
"History of Changes for Study: NCT02690558. Phase 2 Study of Pembrolizumab in Combination With Gemcitabine and Cisplatin as Neoadjuvant Therapy," NCT02690558, Mar. 10, Mar. 2017, Retrieved from the Internet: URL:https://clinicaltrials.gov/ct2/history/NCT02690558?V_4=View#StudyPageTop, 5 pages.
Abdiche et al., "Assessing kinetic and epitopic diversity across orthogonal monoclonal antibody generation platforms." mABs (Feb.-Mar. 2016); 8(2):264-277.
Ahmadzadeh, M. et al., "Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired," Blood. Aug. 20, 2009;114(8):1537-1544. doi: 10.1182/blood-2008-12-195792. Epub May 7, 2009.
Araki, K. et al., "Programmed cell death 1-directed immunotherapy for enhancing T-cell function," Cold Spring Harbor Symposia on Quantitative Biology, vol. LXXVIII, 239-247 (2013).
Arlauckas, S.P. et al., "In vivo imaging reveals a tumor-associated macrophage-mediated resistance pathway in anti-PD-1 therapy," Sci. Transl. Med., 9, eaal3604 (May 2017).
Balar, A. et al., "Pembrolizumab (pembro) as first-line therapy for advanced/unresectable or metastatic urothelial cancer: Preliminary results from the phase 2 KEYNOTE-052 study," Annals of Oncology 27 (Supplement 6): vi552-vi587, 2016.
Balbach, S. et al., "Pharmaceutical evaluation of early development candidates ‘the 100 mg-approach,’" International Journal of Pharmaceutics, 275 (2004), pp. 1-12.
Bellmunt, J. et al., "Keynote-045: open-label, phase II study of pembrolizumab versus investigator's choice of paclitaxel, docetaxel, or vinflunine for previously treated advanced urothelial cancer," Journal for ImmunoTherapy of Cancer, 2016, 4(Suppl. 2):91, Presented at 31st Society for Immunotherapy of Cancer Annual Meeting, Nov. 9-13, 2016, National Harbor, MD, 1 page.
Bellmunt, J. et al., "Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma," N. Engl. J. Med., vol. 376, No. 11, Mar. 2017, pp. 1015-1026.
Bellmunt, J. et al., "Randomized Phase III Study Comparing Paclitaxel/Cisplatin/Gemcitabine and Gemcitabine/Cisplatin in Patients with Locally Advanced or Metastatic Urothelial Cancer Without Prior Systemic Therapy: EORTC Intergroup Study 30987," J Clin Oncol., Apr. 1, 2012;30(10):1107-1113.
Berger, R. et al., "Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies," Clinical Cancer Research, 14(10):3044-3051 (May 2008).
Boyd et al., "Deep sequencing and human antibody repertoire analysis." Current Opinion in Immunology (Jun. 2016); 40: 103-109. Epub Apr. 8, 2016.
Bradshaw, J. M., "The Src, Syk, and Tec family kinases: Distinct types of molecular switches," Cell. Signalling, 22:1175-1184 (2010).
Brahmer, J. R. et al., "Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates," J Clin Oncol. Jul. 1, 2010;28(19):3167-75.
Brahmer, J. R. et al., "Safety and Activity of Anti-PD-L1 Antibody in Patients with Advanced Cancer," N Engl J Med. (Jun. 28, 2012), 366(26):2455-2465.
Brand, F-X et al., "Prospect for anti-HER2 receptor therapy in breast cancer," Anticancer Research, 26:463-470 (2006).
Caira, E. D. et al., "Crystalline polymorphism of organic compounds," Topics in Current Chemistry, vol. 198, 1998, pp. 163-208.
Cartigny, D. et al., "General Asymmetric Hydrogenation of 2-Alkyl- and 2-Aryl-Substituted Quinoxaline Derivatives Catalyzed by Iridium-Difluorphos: Unusual Halide Effect and Synthetic Application," J. Org. Chem., Apr. 2012, vol. 77, No. 10, pp. 4544-4556.
Casset, F. et al., "A peptide mimetic of an anti-CD4 monoclonal antibody by rational design," Biochemical and Biophysical Research Communications, 307:198-205 (2003).
Clynes, R. A. et al., "Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets," Nat. Med. 6(4):443-446 (Apr. 2000).
Conley, M. E. et al., "Primary B Cell Immunodeficiencies: Comparisons and Contrasts," Annu. Rev. Immunol., 27:199-227 (2009).
Conroy, et al., "Antibodies: From novel repertoires to defining and refining the structure of biologically important targets," Methods (Mar. 2017); 116:12-22. Epub Jan. 11, 2017.
Dahan, R. et al., "FcRs Modulate the Anti-tumor Activity of Antibodies Targeting the PD-1/PD-L1 Axis," Cancer Cell (Sep. 2015), 28(3):285-95. doi: 10.1016/j.ccell.2015.08.004.
Damschroder et al., "Analysis of human and primate CD2 molecules by protein sequence and epitope mapping with anti-human CD2 antibodies," Mol Immunol. (Aug. 2004), 41(10):985-1000.
Davis, R. E. et al., "Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma," Nature, 463:88-92 (2010).
De Pascalis, R. et al., "Grafting of‘Abbreviated’ Complementarity-Determining Regions Containing Specificity-Determining Residues Essential for Ligand Contact to Engineer a Less Immunogenic Humanized Monoclonal Antibody," The Journal of Immunology, vol. 169, pp. 3076-3084 (2002).
De Toni, E. N. et al., "Tapering of Immunosuppression and Sustained Treatment With Nivolumab in a Liver Transplant Recipient," Gastroenterology, May 2017;152(6):1631-1633. doi: 10.1053/j.gastro.2017.01.063.
Desai, J. et al., "Updated safety, efficacy, and pharmacokinetics (PK) results from thephase I study of BGB-A317, an anti-programmed death-1 (PD-1) mAb in patients with advanced solid tumors," J. Immunother. Cancer, 2016; 4(Suppl 1):P154, 2 pages.
Dorfman, D. M. et al., "Programmed death-1 (PD-1) is a marker of germinal center-associated T Cells and angioimmunoblastic T-cell lymphoma," American Journal of Surgical Pathology, 30(7):802-810 (Jul. 2006).
El-Khoueiry, A. B. et al., "Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial," Lancet, Jun. 24, 2017;389(10088):2492-2502. doi: 10.1016/S0140-6736(17)31046-2.
European Search Report for European Application No. 16167542.6, dated Nov. 14, 2016, 5 pages.
Extended European Search Report for European Application No. 14787642.9, dated Jan. 26, 2016, 5 pages.
Extended European Search Report for European Application No. 15815646.3, dated Dec. 21, 2017, 10 pages.
Extended European Search Report for European Application No. 17841107.0, dated Feb. 21, 2020, 12 pages.
Extended European Search Report for European Application No. 17841172.4, dated Mar. 5, 2020, 6 pages.
Extended European Search Report for European Application No. 18744173.8, dated Oct. 21, 2020, 12 pages.
Extended European Search Report for European Application No. 18823691.3, dated Feb. 22, 2021, 9 pages.
Ferrara et al., "Recombinant renewable polyclonal antibodies." mABs (2015); 7(1):32-41.
Fuller, M. J. et al., "Immunotherapy of chronic hepatitis C virus infection with antibodies against programmed cell death-1 (PD-1)," Proceedings of the National Academy of Sciences, 110(37):15001-15006 (Sep. 2013).
Galsky, M. D. et al., "Effectiveness of Adjuvant Chemotherapy for Locally Advanced Bladder Cancer," J Clin Oncol. Mar. 10, 2016;34(8):825-832.
Gelderman, K. A. et al., "Complement function in mAb-mediated cancer immunotherapy," Trends in Immunology, 25(3):158-164 (Mar. 2004).
Gurcan, H. M. et al., "A review of the current use of rituximab in autoimmune diseases," Int. Immunopharmacol., 9:10-25 (2009).
Hackam, D. G. et al., "Translation of research evidence from animals to humans," JAMA, 296(14):1731-1732 (2006).
Hamid, O. et al., "Safety and tumor responses with lambrolizumab (Anti-PD-1) in melanoma," New England Journal of Medicine, 369(2):134-144 (Jul. 2013).
Hirayama, Y., "Handbook for organic compound crystal—Principle and know-how," 2008, 28 pages.
Humphries, L. A. et al., "Tec Kinases Mediate Sustained Calcium Influx via Site-specific Tyrosine Phosphorylation of the Phospholipase Cγ Src Homology 2-Src Homology 3 Linker," J. Biol.Chem. 279(36):37651-37661 (2004).
International Search Report and Written Opinion for International Application No. PCT/CN2013/083467, dated Jun. 16, 2014, 9 pages.
International Search Report and Written Opinion for International Application No. PCT/CN2014/075943, dated Jul. 18, 2014, 10 pages.
International Search Report and Written Opinion for International Application No. PCT/CN2015/083066, dated Sep. 24, 2015, 8 pages.
International Search Report and Written Opinion for International Application No. PCT/CN2017/098023, dated Nov. 16, 2017, 10 pages.
International Search Report and Written Opinion for International Application No. PCT/CN2018/074108, dated Apr. 23, 2018, 12 pages.
International Search Report and Written Opinion for International Application No. PCT/CN2018/092827, dated Sep. 29, 2018, 14 pages.
International Search Report and Written Opinion for International Application No. PCT/CN2018/100145, dated Nov. 14, 2018, 8 pages.
International Search Report and Written Opinion for International Application No. PCT/IB2017/054955, dated Sep. 10, 2018, 16 pages.
International Search Report and Written Opinion for International Application No. PCT/US2018/063068, dated Feb. 27, 2019, 8 pages.
International Search Report for International Application No. PCT/US2019/017313, dated Jun. 25, 2019, 16 pages.
InvivoGen Insight, "IgG-Fc Engineering for Therapeutic Use," Apr./May 2006, 4 pages.
James, L. K. et al., "Potential Mechanisms for IgG4 Inhibition of Immediate Hypersensitivity Reactions," Curr Allergy Asthma Rep. 2016; 16: 23. Published online Feb. 18, 2016. doi: 10.1007/s11882-016-0600-2.
Jenkins, S. M. et al., "Substituent variation in azabicyclic triazole- and tetrazole-based muscarinic receptor ligands," J. Med. Chem., 35(13):2392-2406 (1992).
Jiao, Y. et al., "Advances in the research of the anti-cancer agent—Raf kinase inhibitor," Strait Pharmaceutical Journal, vol. 19, No. 8, 2007, pp. 1-5 (with English Abstract).
Jie, L., "Deuterated Drugs Progress," Chemical Engineering Design Communication Medicine and Chemical Industry, 2016, vol. 42 (4), pp. 199.
Jordan, V. C., "Tamoxifen: A most unlikely pioneering medicine," Nature Reviews: Drug Discovery, 2:205-213 (2003).
Kersseboom, R. et al., "Constitutive activation of Bruton's tyrosine kinase induces the formation of autoreactive IgM plasma cells," Eur. J. Immunol. 40:2643-2654, 2010.
Khan et al., "Cross-neutralizing anti-HIV-1 human single chain variable fragments (scFvs) against CD4 binding site and N332 glycan identified from a recombinant phage library." Scientific Reports (2017); Article No. 45163, 12 pages.
Khan, W. N., "Regulation of B lymphocyte development and activation by Bruton's tyrosine kinase," Immunol. Res., 23(2/3):147-156 (2001).
Kim, K.-H. et al., "Imidazo[1,5-a]quinoxalines as irreversible BTK inhibitors for the treatment of rheumatoid arthritis," Bioorg. Med. Chem. Lett., 21:6258-6263 (2011).
Konitzer et al., "Generation of a highly diverse panel of antagonistic chicken monoclonal antibodies against the GIP receptor," mABs (Apr. 2017); 9(3):536-549. Epub Jan. 5, 2017.
Kudo, M., "Immune Checkpoint Inhibition in Hepatocellular Carcinoma: Basics and Ongoing Clinical Trials," Oncology, 2017;92 Suppl 1:50-62. doi: 10.1159/000451016. Epub Feb. 2, 2017.
Kudo, M., Immune Checkpoint Blockade in Hepatocellular Carcinoma: 2017 Update, Liver Cancer, Nov. 2016; 6(1):1-12.
Lee et al., "Molecular-level analysis of the serum antibody repertoire in young adults before and after seasonal influenza vaccination." Nat Med. (Dec. 2016); 22(12):1456-1464. Epub Nov. 7, 2016.
Li, N. et al., "BGB-3111 is a novel and highly selective Bruton's tyrosine kinase (BTK) inhibitor," Cancer Center, vol. 75, No. 15, Supp. 1, Abstract No. 2597, 106th Annual Meeting of the American Association for Cancer Research, AACR 2015, Philadelphia, PA, United States, Aug. 2015, 2 pages.
Lou, Y. et al., "Bruton's tyrosine kinase inhibitors: Approaches to potent and selective inhibition, preclinical and clinical evaluation for inflammatory diseases and B cell malignancies," J. Med. Chem., 55(10):4539-4550 (2012).
Lund, J. et al., "Multiple interactions of IgG with its core oligosaccharide can modulate recognition by complement and human Fc gamma receptor I and influence the synthesis of its oligosaccharide chains," J Immunol., Dec. 1, 1996, 157(11):4963-4969.
Luo, J. et al., "Modern Physical Pharmaceutics Theory and Practice," Shang Hai Science and Technology Literature Publishing House, Apr. 2005, pp. 293-295.
MedChemExpress, "Zanubrutinib," Product Data Sheet, Retrieved from the Internet: www.medchemexpress.com, Retrieved Aug. 17, 2021, 2 pages.
Mohamed, A. J. et al., "Bruton's tyrosine kinase (Btk): function, regulation, and transformation with special emphasis on the PH domain," Immunol. Rev., 228:58-73 (2009).
Pan, Z, "Bruton's tyrosine kinase as a drug discovery target," Drug News Perspect, 21(7):357-362 (2008).
Panka, D. J. et al., "Variable region framework differences result in decreased or increased affinity of variant anti-digoxin antibodies," May 1988, Proc. Natl. Acad. Sci. USA, 85:3080-3084.
Parola et al., "Integrating high-throughput screening and sequencing for monoclonal antibody discovery and engineering." Immunology (Jan. 2018); 153(1):31-41. Epub Oct. 30, 2017.
Paul, W. E. (Ed.), Chap. 9, Structure and Function of Immunoglobulins, In: Fundamental Immunology, Third Edition, pp. 292-295, 1993.
Plimack, E. R. et al., "Pembrolizumab (MK-3475) for advanced urothelial cancer: Updated results and biomarker analysis from KEYNOTE-012," J. Clin. Oncol., vol. 33, Issue 15 Suppl., May 2015, Abstract 4502, 2 pages.
Presta, L. G. et al., "Engineering therapeutic antibodies for improved function," Biochemical Society Transactions (2002) vol. 30, Part 4, pp. 487-490.
Rokosz, L. L. et al., "Kinase inhibitors as drugs for chronic inflammatory and immunological diseases: progress and challenges," Expert Opin. Ther. Targets, 12(7):883-903 (2008).
Rudikoff, S. et al., "Single amino acid substitution altering antigen-binding specificity," Proc. Natl. Acad. Sci. USA, 79:1979-1983 (Mar. 1982).
Samonakis, D. N. et al., "Systemic treatment for hepatocellular carcinoma: Still unmet expectations," World J Hepatol., Jan. 2017; 9(2):80-90.
Sequence Alignment, 2014, 1 page.
Sharma, P. et al., "Efficacy and safety of nivolumab monotherapy in metastatic urothelial cancer (mUC): Results from the phase I/II CheckMate 032 study," J. Clin. Oncol., vol. 34, No. 15 Suppl., May 2016, pp. 4501.
Sharma, P. et al., "Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, open-label, two-stage, multi-arm, phase 1/2 trial," Lancet Oncol., Nov. 2016, vol. 17, No. 11, pp. 1590-1598.
Sheehan et al., "Phage and Yeast Display." Microbial. Spectr. (2015); 3(1):AID-0028-2014; 17 pages.
Shields, R. L. et al., "High Resolution Mapping of the Binding Site on Human IgG1 for FcRI, FcRII, FcRIII, and FcRn and Design of IgG1 Variants with Improved Binding to the FcR," The Journal of Biological Chemistry, 276(9):6591-6604 (2001).
Shioji, Y., "Production Technology of Solid Preparations," Tokyo, CMC Publication, Jan. 27, 2003, Popular Edition, pp. 9 and 12-13.
Singhal, D. et al., "Drug polymorphism and dosage form design: a practical perspective," Advanced Drug Delivery Reviews, 56 (2004) pp. 335-347.
Smith, C. I. E. et al., "Expression of Bruton's Agammaglobulinemia Tyrosine Kinase Gene, BTK, Is Selectively Down-Regulated in T Lymphocytes and Plasma Cells," J. Immunol., 152:557-565 (1994).
Smith, K. G. et al., "FcRIIB in autoimmunity and infection: evolutionary and therapeutic implications," Nat Rev Immunol. May 2010;10(5):328-43.
Stave, J. W. et al., "Antibody and antigen contact residues define epitope and paratope size and structure," The Journal of Immunology, vol. 191, Jan. 2013, pp. 1428-1435.
Strome, S. E. et al., "A Mechanistic Perspective of Monoclonal Antibodies in Cancer Therapy Beyond Target-Related Effects," The Oncologist, 2007; 12:1084-1095.
Supplementary Partial European Search Report for European Application No. 13893636.4, dated Feb. 28, 2017, 13 pages.
Sznol, M. et al., "Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer," Clinical Cancer Research, 19(5):1021-1034 (Mar. 2013).
Takada, N., "Bulk Drug Form Screening and Selection at Drug Discovery Phase," Pharm Stage, Jan. 15, 2007 vol. 6, No. 10, pp. 20-25.
Takayama, T. et al., "Effects of the novel and potent lymphocyte-specific protein tyrosine kinase inhibitor TKM0150 on mixed lymphocyte reaction and contact hypersensitivity in mice," Arzneimittelforschung, 60(5):282-285 (2010).
Takayama, T. et al., "Ring-fused pyrazole derivatives as potent inhibitors of lymphocyte-specific kinase (Lck): Structure, synthesis, and SAR," Bioorganic & Medicinal Chemistry Letters, 20(1):112-116 (Jan. 2010).
Topalian, S. L., "Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer," N. Engl. J. Med., Jun. 2012; vol. 366, No. 26, pp. 2443-2454.
Uckun, F. M. et al., "Bruton's tyrosine kinase as a new therapeutic target," Anti-Cancer Agents in Medicinal Chemistry, 7(6):624-632 (2007).
Van Regenmortel, M. H. V., "Development of a Preventive HIV Vaccine Requires Solving Inverse Problems Which Is Unattainable by Rational Vaccine Design." Front Immunol. (Jan. 2018); 8: 2009. eCollection 2017.
Vetrie, D. et al., "The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases," Nature, 361:226-233 (1993).
Wang, C. et al., "In Vitro Characterization of the Anti-PD-1 Antibody Nivolumab, BMS-936558, and In Vivo Toxicology in Non-Human Primates," Cancer Immunol Res; 2(9):846-856 (Sep. 2014).
Wherry, E. J., "T cell exhaustion," Nature Immunology 12(6):492-499 (2011). Published online May 18, 2011.
Wilson, W. H. et al., "686—The Bruton's Tyrosine Kinase (Btk) Inhibitor, Ibrutinib (PCI-32765), Has Preferential Activity in the ABC Subtype of Relapsed/Refractory De Novo Diffuse Large B-Cell Lymphoma (DLBCL): Interim Results of a Multicenter, Open-Label, Phase2 Study," Poster #686, 54th American Society of Hematology (ASH) annual meeting abstract (Dec. 10, 2012).
Wu, H. et al., "Humanization of a murine monoclonal antibody by simultaneous optimization of framework and CDR residues," Journal of Molecular Biology, 294(1):151-162 (Nov. 1999).
Xu, D. et al., "In vitro characterization of five humanized OKT3 effector function variant antibodies," Cell Immunol. Feb. 25, 2000;200(1):16-26.
Yen, C-J et al., "Preliminary results of a phase 1A/1B study of BGB-A317, an anti-PD-1 monoclonal antibody (mAb), in patients with advanced hepatocellular carcinoma (HCC)," Abstract P-140, Annals of Oncology, vol. 28, Supplement 3, p. 54 (2017).
Zhou et al., "Structural Repertoire of HIV-1-Neutralizing Antibodies Targeting the CD4 Supersite in 14 Donors." Cell (Jun. 2015); 161(6):1280-1292.

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11851437B2 (en) 2016-08-16 2023-12-26 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11884674B2 (en) 2016-08-16 2024-01-30 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra- hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11701357B2 (en) 2016-08-19 2023-07-18 Beigene Switzerland Gmbh Treatment of B cell cancers using a combination comprising Btk inhibitors
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors

Also Published As

Publication number Publication date
CN110461847B (en) 2022-06-07
TWI774726B (en) 2022-08-21
EP3573989A1 (en) 2019-12-04
CN110461847A (en) 2019-11-15
WO2018137681A1 (en) 2018-08-02
EP3573989A4 (en) 2020-11-18
TW201831488A (en) 2018-09-01
US20210332049A1 (en) 2021-10-28

Similar Documents

Publication Publication Date Title
US11555038B2 (en) Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11851437B2 (en) Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
JP6850922B2 (en) Diazanaphthalene compounds as JAK kinase inhibitors
US20210206757A1 (en) N-substituted-dioxocyclobutenylamino-3-hydroxy-picolinamides useful as ccr6 inhibitors
US20180282310A1 (en) Forms and compositions of biaryl inhibitors of bruton&#39;s tyrosine kinase
US20230141887A1 (en) Crystal form of diazaspiropyran compound
WO2022049253A1 (en) Substituted n-heteroaryl-n-pyridinylacetamides as p2x4 modulators

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: BEIGENE, LTD., CAYMAN ISLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GUO, YUNHANG;YU, DESHENG;WANG, ZHIWEI;SIGNING DATES FROM 20180515 TO 20180518;REEL/FRAME:049830/0832

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE